EP3240569A1 - Anti-cd37-antikörper und verwendungen davon - Google Patents

Anti-cd37-antikörper und verwendungen davon

Info

Publication number
EP3240569A1
EP3240569A1 EP15876093.4A EP15876093A EP3240569A1 EP 3240569 A1 EP3240569 A1 EP 3240569A1 EP 15876093 A EP15876093 A EP 15876093A EP 3240569 A1 EP3240569 A1 EP 3240569A1
Authority
EP
European Patent Office
Prior art keywords
antibody
amino acid
seq
cell
heavy chain
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP15876093.4A
Other languages
English (en)
French (fr)
Other versions
EP3240569A4 (de
Inventor
Aaron Sato
Ryan STAFFORD
Junhao Yang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Celgene Corp
Original Assignee
Celgene Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Celgene Corp filed Critical Celgene Corp
Publication of EP3240569A1 publication Critical patent/EP3240569A1/de
Publication of EP3240569A4 publication Critical patent/EP3240569A4/de
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39541Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against normal tissues, cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/02Preparation of peptides or proteins having a known sequence of two or more amino acids, e.g. glutathione
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • C07K2317/14Specific host cells or culture conditions, e.g. components, pH or temperature
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/734Complement-dependent cytotoxicity [CDC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Definitions

  • anti-CD47 antibodies which specifically bind to CD47 and compositions comprising such antibodies, including pharmaceutical compositions, diagnostic compositions and kits. Also provided are methods of using anti-CD74 antibodies for therapeutic and diagnostic purposes, and methods for making such anti-CD47 antibodies, for example with cell-free (CF) systems.
  • CF cell-free
  • CD47 also known as integrin-associated protein (IAP), ovarian cancer antigen OA3, Rh-related antigen and MER6, is a multi-spanning transmembrane receptor belonging to the immunoglobulin superfamily.
  • SIRPa signal-regulatory-protein a expressed on macrophages interacts with CD47, and this interaction negatively controls effector function of innate immune cells such as host cell phagocytosis.
  • CD47 expression and/or activity have been implicated in a number of diseases and disorders. Accordingly, there exists a need for therapies that target CD47, as well as better methods for making such therapies.
  • antibodies e.g., monoclonal antibodies
  • CD47 e.g., human CD47
  • ECD extracelluar domain
  • anti-CD47 antibody blocks CD47 binding to SIRPa, promotes phagocytosis, has reduced or no Fc effector function (e.g., binding to FcyR, ADCC, or CDC) and/or has little or no agglutination (e.g., hemagglutination) activity.
  • a monoclonal anti-CD47 antibody which specifically binds to human CD47, wherein the anti-CD47 antibody is a variant of a parental antibody, and wherein the anti-CD47 antibody when produced using a cell-free (CF) expression system has a higher antibody expression titer or yield compared to the parental antibody when produced in the CF expression system.
  • anti-CD47 antibodies provided herein which are expressed in a CF system are aglycosylated.
  • a monoclonal anti-CD47 antibody which specifically binds to human CD47 (e.g., SEQ ID NO: 38 or 39), wherein the anti-CD47 antibody, when produced using a cell-free system, has a higher antibody expression titer or yield compared to a parental antibody produced using the cell-free system.
  • the anti-CD47 antibody expression titer or yield is higher by at least 1 fold, at least 2 fold, or at least 3 fold compared to the parental antibody.
  • the anti-CD47 antibody expression titer or yield is higher by at least 25%, 50%, 75%, or 100% compared to the parental antibody.
  • such anti-CD47 antibody is a humanized antibody.
  • the cell-free system comprises using S30 cell-free extract.
  • such cell-free system comprises prokaryotic disulfide bond isomerase DsbC.
  • such anti- CD47 antibody is an IgGi antibody.
  • such anti-CD47 antibody is an IgG 4 antibody.
  • such anti-CD47 antibody is an IgG 4 antibody comprising a S228P amino acid substitution according to the EU numbering index.
  • such anti- CD47 antibody is an IgG 4 antibody comprising a S228P and L235E amino acid substitutions according to the EU numbering index.
  • such parental antibody of an anti-CD47 antibody provided herein comprises a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 1.
  • such parental antibody comprises a light chain variable region comprising the amino acid sequence of SEQ ID NO: 12.
  • such parental antibody comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 2, 3 or 4.
  • an anti-CD47 antibody provided herein comprises (i) a heavy chain variable region comprising complementarity determining region (CDR) 1, 2, and 3 of antibody 2A1; and (ii) a light chain variable region comprising CDR1, CDR2, and CDR3 of antibody 2A1.
  • CDR complementarity determining region
  • an anti-CD47 antibody comprises (i) a heavy chain variable region comprising complementarity determining region (CDR) 1, 2, and 3 comprising amino acid sequences GFNIKDYYLH (SEQ ID NO: 14), WIDPDQGDTE (SEQ ID NO: 15), and NAAYGSSSYPMDY (SEQ ID NO: 16), respectively; and (ii) a light chain variable region comprising CDR1, CDR2, and CDR3 comprising amino acid sequences
  • KASQDIHRYLS SEQ ID NO: 17
  • RANRLVS SEQ ID NO: 18
  • LQYDEFPYT SEQ ID NO: 19
  • anti-CD47 antibody comprising one or more amino acid modifications (e.g., amino acid substitutions) relative to a parental antibody.
  • such one or more amino acid substitutions is in the framework region of the heavy chain variable region or light chain variable region.
  • such anti-CD47 antibody comprises 13 or 14 amino acid modifications (e.g., amino acid substitutions) in the framework region of the heavy chain variable region.
  • such anti-CD47 antibody comprises 1 to 15 amino acid modifications (e.g., amino acid substitutions) in the framework region of the heavy chain variable region.
  • amino acid modifications are conservative amino acid substitutions.
  • a monoclonal anti-CD47 antibody which specifically binds to CD47 (e.g., human CD47 such as SEQ ID NO: 38 or 39) and comprises a heavy chain variable region (V H ) comprising the amino acid sequence:
  • Xi is M or there is no amino acid at position Xi
  • X 2 is an amino acid with hydrophobic side chains such as M or V
  • X 3 is T or P
  • X 4 is S or A
  • X 5 is an amino acid having aliphatic side chains such as A or G
  • X 6 is F or L
  • X 7 is D or G
  • X 8 is an amino acid with hydrophobic side chains such as I or M
  • X 9 is R or T
  • Xi 0 is R or T
  • X u is M or T
  • Xi 2 is S or R
  • X i3 is a negatively charged amino acid such as E or D
  • Xi 4 is an amino acid with hydrophobic side chains such as M or V.
  • the V H of an anti-CD47 antibody provided herein comprises the amino acid sequence of SEQ ID NO: 21. In certain aspects, the V H of an anti-CD47 antibody provided herein comprises the amino acid sequence of SEQ ID NO: 22. In specific aspects, an anti-CD47 antibody provided herein comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 5. In particular aspects, an anti-CD47 antibody provided herein comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 6. In certain aspects, an anti-CD47 antibody provided herein comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 7. In particular aspects, an anti-CD47 antibody provided herein comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 8.
  • an anti-CD47 antibody provided herein comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 9. In certain aspects, an anti-CD47 antibody provided herein comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 10. In particular aspects, an anti-CD47 antibody provided herein comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 11.
  • an anti-CD47 antibody provided herein comprises a light chain comprising the amino acid sequence of SEQ ID NO: 13 or SEQ ID NO: 13 without amino acid M at the N-terminus.
  • a monoclonal anti-CD47 antibody which specifically binds to CD47 (e.g., human CD47 such as SEQ ID NO: 38 or 39), wherein the anti- CD47 antibody does not cause or promote substantial red blood cell depletion, anemia, or both red blood cell depletion and anemia after administration.
  • CD47 e.g., human CD47 such as SEQ ID NO: 38 or 39
  • the anti- CD47 antibody does not cause or promote substantial platelet depletion after administration.
  • such anti-CD47 antibody does not cause or promote substantial agglutination of cells after administration.
  • such anti-CD47 antibody does not cause or promote substantial hemagglutination of red blood cells after administration.
  • such anti-CD47 antibody inhibits (e.g., inhibits by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100%) CD47 from interacting with signal -regulatory-protein a
  • such anti-CD47 antibody promotes phagocytosis, such as macrophage-mediated phagocytosis of a CD47-expressing cell.
  • such anti- CD47 antibody provided herein does not cause or promote a significant level of effector function.
  • an anti-CD47 antibody provided herein when expressed using a cell-free system, exhibits lower (e.g., lower by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%) binding affinity, or does not bind, to an FcyR compared to when expressed using CHO cells.
  • such lower binding affinity is at least 1 log lower or at least 2 log lower.
  • the FcyR is FcyRI, FcyRIIA R131, FcyRIIA H131, FcyRIIB, or FcyRIIIA VI 58.
  • an anti-CD47 antibody provided herein is aglycosylated or has less (e.g., at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% less) glycosylation when expressed using the cell free system compared to when expressed in CHO cells.
  • a monoclonal anti-CD47 antibody which specifically binds to CD47 (e.g., human CD47 such as SEQ ID NO: 38 or 39), wherein the anti- CD47 antibody (i) promotes phagocytosis such as macrophage-mediated phagocytosis of a CD47-expressing cell; (ii) does not cause or promote a significant level of hemagglutination of red blood cells after administration; (iii) does not cause or promote a significant level of ADCC or CDC; and/or (iv) exhibits lower (e.g., lower by at least about 10%, 20%, 30%, 40%, 50%, 60%), 70%), 80%), 90%) binding affinity, or does not bind, to an FcyR compared to when expressed using CHO cells or compared to a parental antibody.
  • CD47 e.g., human CD47 such as SEQ ID NO: 38 or 39
  • the anti- CD47 antibody promotes phagocytosis such as macrophage-mediated phagocytosis
  • an anti-CD47 antibody provided herein is a bispecific antibody.
  • an anti-CD47 antibody provided herein is conjugated to an agent.
  • the agent is a label or a toxin.
  • a pharmaceutical composition comprising an effective amount of an anti-CD47 antibody provided herein or an antigen-binding fragment thereof.
  • the pharmaceutifcal composition provided herein further comprising a pharmaceutically acceptable carrier.
  • a polynucleotide comprising a nucleotide sequence encoding a VH chain region, a VL chain region, or both a VL chain region and a VH chain region, of an anti-CD47 antibody described herein.
  • a polynucleotide comprising a nucleotide sequence encoding a heavy chain, a light chain, or both heavy chain and a light chain of an anti-CD47 antibody described herein.
  • such polynucleotide comprises a nucleotide sequence of any one of SEQ ID NOs: 26-32 encoding a heavy chain.
  • such polynucleotide comprises a nucleotide sequence of SEQ ID NO: 33 encoding a light chain.
  • a population of polynucleotides comprising (i) a first polynucleotide comprising nucleotide sequences encoding a VH or a heavy chain of an anti-CD47 antibody described herein, and (ii) a second polypeptide comprising nucleotide sequences encoding a VL or a light chain of an anti-CD47 antibody described herein.
  • first polynucleotide is operably linked to a first promoter
  • such second polynucleotide is operably linked to a second promoter.
  • a vector comprising one or more
  • a population of vectors comprising (i) a first vector comprising nucleotide sequences encoding a VH or a heavy chain of an anti-CD47 antibody described herien, and (ii) a second vector comprising nucleotide sequences encoding a VL or a light chain of an anti-CD47 antibody described herein.
  • compositions for cell-free protein expression comprising a cell-free extract and one or more polynucleotides or vectors described herein.
  • the composition further comprising S30 cell-free extract.
  • the composition provided herein further comprises prokaryotic disulfide bond isomerase DsbC.
  • provided herein is a method of treating cancer, wherein the method comprises administering an anti-CD47 antibody described herein to a subject in need thereof in an amount sufficient to treat the cancer in the subject.
  • a method of alleviating a symptom of a cancer comprising administering an anti-CD47 antibody described herein to a subject in need thereof in an amount sufficient to alleviate one or more symptoms of the cancer in the subject.
  • such method provided herein further comprises administering radiation or chemotherapy.
  • such method provided herein further comprises administering another anti-cancer agent.
  • the cancer is a hematological cancer.
  • the cancer is a solid cancer.
  • the cancer is multiple myeloma, non-Hodgkin's lymphoma, acute myeloid leukemia (AML), breast cancer, bladder cancer, non-small cell lung cancer/carcinoma, hepatocellular carcinoma (HCC), sarcoma, or head and neck cancer.
  • provided herein is an isolated cell comprising one or more polynucleotides or vectors described herein.
  • an isolated cell comprising a population of polynucleotides or vectors described herein.
  • a population of host cells comprising (i) a first host cell comprising a polynucleotide comprising nucleotide sequences encoding a VH or a heavy chain of an anti-CD47 antibody described herein, and (ii) a second host cell comprising a polynucleotide comprising nucleotide sequences encoding a VL or a light chain of an anti-CD47 antibody described herein.
  • an anti-CD47 antibody comprising expressing an anti-CD47 antibody described herein with a composition for cell-free protein expression described herein. In a certain aspect, such method further comprises purifying the anti-CD47 antibody.
  • an anti-CD47 antibody comprising expressing the anti-CD47 antibody with a cell described herein. In a certain aspect, such method further comprises purifying the anti-CD47 antibody.
  • Figure 1A depicts the autoradiagram of anti-CD47 antibodies expressed with a cell- free (CF) system.
  • Samples 1-10 correspond to CF-expressed anti-CD47 IgGl (1), IgGl-5m (2), IgGl-13m (3), IgGl-13mZ (4), IgG4P (5), IgG4P-5m (6), IgG4P-13m (7), IgG4PE (8), IgG4PE- 5m (9), and IgG4PE-13m (10) antibodies, respectively.
  • Figure IB depicts a graph showing anti-CD47 antibody titers (mg/L) obtained from aCF expression system.
  • Samples 1-10 correspond to CF-expressed anti-CD47 IgGl (1), IgGl- 5m (2), IgGl-13m (3), IgGl-13mZ (4), IgG4P (5), IgG4P-5m (6), IgG4P-13m (7), IgG4PE (8), IgG4PE-5m (9), and IgG4PE-13m (10) antibodies, respectively.
  • Figures 2A-2F depict individual sensorgrams from Biacore analysis of anti-CD47 IgGl -5m (2A), IgGl-13m (2B), IgGl-13mZ (2C), IgG4P-5m (2D), IgG4PE-5m (2E), and control antibody (2F).
  • Figures 3A-3C depict graphs plotting specific heat capacity (kcal/mol/°C) versus temperature (°C) from thermostability analysis using Differential Scanning Calorimetry (DSC) for anti-CD47 IgGl-13mZ (3A), IgGl-13m (3B), and IgGl-5m (3C) antibodies.
  • Figure 4 depicts a graph plotting anti-CD47 antibody plasma concentration ⁇ g/mL) versus time (hours) from pharmacokinetic studies with anti-CD47 IgG4-PE antibody produced with CHO cells and anti-CD47 IgGl and IgGl -5m antibodies produced by the CF expression system.
  • Figure 5 depicts a graph plotting tumor volume (mm 3 ) versus days after RPMI8226 tumor cell inoculation from in vivo mouse tumor xenograft studies using anti-CD47 IgGl -5m antibodies produced by the CF expression system at doses of 1 mg/kg, 0.3 mg/kg, and 0.1 mg/kg (qwx3).
  • antibodies ⁇ e.g., monoclonal antibodies
  • antigen-binding fragments thereof that specifically bind to CD47 (e.g., human CD47).
  • CD47 e.g., human CD47
  • anti-CD47 antibody blocks CD47 binding to SIRPa, promotes
  • phagocytosis has reduced or no Fc effector function (e.g., binding to FcyR, ADCC, or CDC) and/or has little or no agglutination (e.g., hemagglutination) activity.
  • Fc effector function e.g., binding to FcyR, ADCC, or CDC
  • agglutination e.g., hemagglutination
  • a monoclonal anti-CD47 antibody which specifically binds to human CD47, wherein the anti-CD47 antibody is a variant of a parental antibody, and wherein the anti-CD47 antibody when produced using a cell-free (CF) expression system has a higher antibody expression titer or yield compared to the parental antibody when expressed in the CF system.
  • anti-CD47 antibodies provided herein which are expressed in a CF system are aglycosylated.
  • CD47 or "integrin-associated protein” or “IAP” or “ovarian cancer antigen” or “OA3” or “Rh-related antigen” or “MER6” can be used
  • amino acid sequence of an exemplary human CD47 excluding the signal sequence is provided below.
  • red blood cell(s) and erythrocyte(s) are synonymous and used interchangeably herein.
  • agglutination refers to cellular clumping
  • hemagglutination refers to clumping of a specific subset of cells, i.e., red blood cells.
  • hemagglutination is a type of agglutination.
  • antibodies which specifically bind to CD47 e.g., human CD47.
  • anti-CD47 antibodies comprising modifications in one or more amino acid residues (e.g., 5-13 amino acid substitutions in the framework region of the heavy chain variable region) that surprisingly allow for better production in a cell-free (CF) expression sytem than the parental antibody without the modifications.
  • CF cell-free
  • anti-CD47 antibodies inhibit SIRPa interaction with CD47, are aglycosylated, promote phagocytosis, either in vivo or in vitro or both, have antitumor activity (e.g., without promoting agglutination, such as hemagglutination), and/or have low or no Fc effector function (e.g., binding to an FcyR, ADCC, or CDC).
  • antibodies or antigen-binding fragments described herein can comprise sequences that do not naturally exist within the antibody germline repertoire of an animal or mammal ⁇ e.g., human) in vivo.
  • the terms "about” or “approximately” mean within plus or minus 10% of a given value or range. In instances where an integer is required, the terms mean within plus or minus 10% of a given value or range, rounded either up or down to the nearest integer.
  • antibody and “immunoglobulin” and “Ig” are terms of art and can be used interchangeably herein and refer to a molecule with an antigen binding site that specifically binds an antigen.
  • Antibodies can include, for example, monoclonal antibodies, recombinantly produced antibodies, monospecific antibodies, multispecific antibodies (including bispecific antibodies), human antibodies, humanized antibodies, murine antibodies (e.g., mouse or rat antibodies), chimeric antibodies, synthetic antibodies, and tetrameric antibodies comprising two heavy chain and two light chain molecules.
  • antibodies can include, but are not limited to an antibody light chain monomer, an antibody heavy chain monomer, an antibody light chain dimer, an antibody heavy chain dimer, an antibody light chain- antibody heavy chain pair, intrabodies, heteroconjugate antibodies, single domain antibodies, and monovalent antibodies.
  • antibodies can include antigen-binding fragments or epitope binding fragments such as, but not limited to, single chain antibodies or single-chain Fvs (scFv) (e.g., including monospecific, bispecific, etc.), camelized antibodies, affybodies, Fab fragments, F(ab') fragments, F(ab') 2 fragments, and disulfide-linked Fvs (sdFv).
  • scFv single chain antibodies or single-chain Fvs
  • sdFv single-chain Fvs
  • antibodies described herein refer to polyclonal antibody populations.
  • Antibodies can be of any type (e.g., IgG, IgE, IgM, IgD, IgA or IgY), any class, (e.g., IgGi, IgG 2 , IgG 3 , IgG 4 , IgAi or IgA 2 ), or any subclass (e.g., IgG 2a or IgG 2b ) of immunoglobulin molecule.
  • antibodies described herein are IgG antibodies, or a class (e.g., human IgGi, IgG 2 , IgG 3 or IgG 4 ) or subclass thereof.
  • antibodies described herein are IgGi antibodies (e.g., human IgGi) or a subclass thereof. In certain embodiments, IgGi antibodies described herein comprise one or more amino acid substitutions and/or deletions in the constant region. In certain embodiments, antibodies described herein are IgG 4 antibodies (e.g., human IgG 4 ) or a subclass thereof. In certain embodiments, IgG 4 antibodies described herein comprise one or more amino acid substitutions and/or deletions in the constant region.
  • an "antigen” is a moiety or molecule that contains an epitope to which an antibody can specifically bind. As such, an antigen is also specifically bound by an antibody.
  • an “epitope” is a term in the art and refers to a localized region of an antigen to which an antibody can specifically bind.
  • An epitope can be a linear epitope or a conformational, non-linear, or discontinuous, epitope.
  • an epitope in the case of a polypeptide antigen, for example, can be contiguous amino acids of the polypeptide (a "linear” epitope) or an epitope can comprise amino acids from two or more non-contiguous regions of the polypeptide (a "conformational,” “non-linear” or “discontinuous” epitope). It will be appreciated by one of skill in the art that, in general, a linear epitope may or may not be dependent on secondary, tertiary, or quaternary structure.
  • the terms “immunospecifically binds,” “immunospecifically recognizes,” “specifically binds,” and “specifically recognizes” are analogous terms in the context of antibodies and refer to molecules that bind to an antigen/epitope as such binding is understood by one skilled in the art.
  • a molecule e.g., an antibody
  • that specifically binds to an antigen may bind to other peptides or polypeptides, generally with lower affinity as determined by, e.g., immunoassays, surface plasm on resonance assays, for example, BiacoreTM, KinExA platform (Sapidyne Instruments, Boise, ID), or other assays known in the art.
  • molecules that specifically bind to an antigen bind to the antigen with a K a that is at least 2 logs, 2.5 logs, 3 logs, 4 logs or greater than the K a W h en the molecules bind to another antigen.
  • molecules that specifically bind to an antigen do not cross react with other proteins.
  • molecules that specifically bind to an antigen do not cross react with other non-CD47 proteins.
  • the term "monoclonal antibody” is a well known term of art that refers to an antibody obtained from a population of homogenous or substantially homogeneous antibodies.
  • the term “monoclonal” is not limited to any particular method for making the antibody.
  • a population of monoclonal antibodies can be generated by cells, a population of cells, or a cell line.
  • a “monoclonal antibody,” as used herein is an antibody produced by a single cell or cell line wherein the antibody
  • a monoclonal antibody can be a chimeric antibody or a humanized antibody.
  • a monoclonal antibody is a monovalent antibody or multivalent (e.g., bivalent) antibody.
  • non-natural amino acid refers to an amino acid that is not a proteinogenic amino acid, or a post-translationally modified variant thereof. In particular, the term refers to an amino acid that is not one of the 20 common amino acids or pyrrolysine or selenocysteine, or post-translationally modified variants thereof.
  • polyclonal antibodies refers to an antibody population that includes a variety of different antibodies that immunospecifically bind to the same and/or to different epitopes within an antigen or antigens.
  • variable region refers to a portion of an antibody, generally, a portion of an antibody light or heavy chain, typically about the amino- terminal 110 to 120 amino acids in a mature heavy chain and about the amino-terminal 90 to 100 amino acids in a mature light chain.
  • Variable regions comprise complementarity determining regions (CDRs) flanked by framework regions (FRs).
  • CDRs complementarity determining regions
  • FRs framework regions
  • spacial orientation of CDRs and FRs are as follows, in an N-terminal to C-terminal direction: FR1-CDR1-FR2-CDR2-FR3- CDR3-FR4.
  • variable region is a human variable region.
  • variable region comprises murine ⁇ e.g., mouse or rat) CDRs and human framework regions (FRs).
  • variable region is a primate ⁇ e.g., human or non-human primate) variable region.
  • variable region comprises murine ⁇ e.g., mouse or rat) CDRs and primate ⁇ e.g., human or non-human primate) framework regions (FRs).
  • FRs framework regions
  • a variable region described herein is obtained from assembling two or more fragments of human sequences into a composite human sequence.
  • the CDRs of an antibody can be determined according to (i) the Kabat numbering system (Kabat et al. (197 ) Ann. NY Acad. Sci. 190:382-391 and, Kabat et al. (1991) Sequences of Proteins of Immunological Interest Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242); or (ii) the Chothia numbering scheme, which will be referred to herein as the "Chothia CDRs" (see, e.g., Chothia and Lesk, 1987, J. Mol. Biol., 196:901-917; Al-Lazikani et al., 1997, J. Mol.
  • CDRs within an antibody heavy chain molecule are typically present at amino acid positions 31 to 35, which optionally can include one or two additional amino acids, following 35 (referred to in the Kabat numbering scheme as 35 A and 35B) (CDRl), amino acid positions 50 to 65 (CDR2), and amino acid positions 95 to 102 (CDR3).
  • CDRs within an antibody light chain molecule are typically present at amino acid positions 24 to 34 (CDRl), amino acid positions 50 to 56 (CDR2), and amino acid positions 89 to 97 (CDR3).
  • the actual linear amino acid sequence of the antibody variable domain can contain fewer or additional amino acids due to a shortening or lengthening of a FR and/or CDR and, as such, an amino acid's Kabat number is not necessarily the same as its linear amino acid number.
  • Antibodies provided herein can be of any type ⁇ e.g., IgG, IgE, IgM, IgD, IgA or IgY), any class, ⁇ e.g., IgGi, IgG 2 , IgG 3 , IgG 4 , IgAi or IgA 2 ), or any subclass ⁇ e.g., IgG 2a or IgG 2 , or a mixture thereof) of immunoglobulin molecule.
  • antibodies described herein are IgG antibodies ⁇ e.g., human IgG), or a class ⁇ e.g., human IgG 1; IgG 2 , IgG 3 or IgG 4 ) or subclass thereof.
  • an antibody comprising an antibody light chain and heavy chain, e.g., a separate light chain and heavy chain.
  • the light chain of an antibody described herein is a kappa ( ⁇ ) light chain.
  • the light chain of an antibody described herein is a lambda ( ⁇ ) light chain.
  • light chain is a mixed sequence, e.g., the variable portion of the light chain comprises kappa light chain sequences and the constant region of the light chain comprises lambda light chain sequences, or vice versa.
  • the light chain of an antibody described herein is a human kappa light chain or a human lambda light chain.
  • Non-limiting examples of human constant region sequences have been described in the art, e.g., see U.S. Patent No. 5,693,780 and Kabat et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91- 3242.
  • an antibody e.g. a monoclonal antibody, which specifically binds to human CD47, wherein such an anti-CD47 antibody is a variant of a parental anti-CD47 antibody, wherein the anti-CD47 antibody, when produced using a cell-free (CF) expression system, has a higher antibody expression titer or yield compared to that of the parental anti-CD47 antibody when expressed in the CF system, and wherein the anti-CD47 antibody comprises one or more amino acid modifications, for example, 1-15 amino acid modifications, relative to the the parental anti-CD47 antibody.
  • CF cell-free
  • the one or more amino acid modifications are within the heavy chain or VH (e.g., SEQ ID NO: 1). In a particular aspect, the one or more amino acid modifications, for example, 1-15 amino acid modifications, are within the framework region of a VH (e.g., SEQ ID NO: 1).
  • the anti-CD47 antibody provided herein which is a variant of a parental anti-CD47 antibody comprising the CDRs (e.g., Kabat CDRs) of the parental anti-CD47 antibody.
  • an antibody e.g. a monoclonal antibody, which specifically binds to human CD47, wherein such an anti-CD47 antibody is a variant of a parental anti-CD47 antibody, wherein the anti-CD47 antibody, when produced using a cell-free (CF) expression system, has a higher antibody expression titer or yield compared to that of the parental anti-CD47 antibody when expressed in the CF system, and wherein the anti-CD47 antibody comprising one or more amino acid modifications, for example, 1-15 amino acid modifications, relative to the the parental anti-CD47 antibody.
  • CF cell-free
  • the one or more amino acid modifications are within the heavy chain or VH (e.g., SEQ ID NO: 1).
  • the one or more amino acid modifications, for example, 5, 10, 13 or 14 amino acid modifications are within the framework region of a VH (e.g., SEQ ID NO: 1).
  • the one or more amino acid modifications, for example, 5, 13 or 14 amino acid modifications are within the framework region of a VH (e.g., SEQ ID NO: 1).
  • the anti-CD47 antibody provided herein which is a variant of a parental anti-CD47 antibody comprising the CDRs (e.g., Kabat CDRs) of the parental anti-CD47 antibody.
  • such anti-CD47 antibody is an IgGl, IgG2, IgG3, or IgG4 isotype antibody.
  • such anti-CD47 antibody is an IgGl isotype antibody.
  • such anti-CD47 antibody is an IgGl Z allotype isotype antibody.
  • such anti-CD47 antibody is an IgG4, such as an IgG4P or IgG4PE, isotype antibody.
  • an antibody e.g. a monoclonal antibody, which specifically binds to human CD47, wherein such an anti-CD47 antibody is a variant of a parental anti-CD47 antibody, wherein the anti-CD47 antibody, when produced using a cell-free (CF) expression system, has a higher antibody expression titer or yield compared to that of the parental anti-CD47 antibody when expressed in the CF system.
  • the parental anti-CD47 antibody is antibody AB6.12 (see, e.g., U.S. Application Publication No. US 2014/0140989 Al, which is incorporated herein by reference in its entirety).
  • the anti-CD47 antibody provided herein is a variant of parental antibody AB6.12, and comprises the CDRs (e.g., Kabat CDRs) of parental antibody AB6.12, for example SEQ ID NOs: 14-19.
  • such anti-CD47 antibody is an IgGl, IgG2, IgG3, or IgG4 isotype antibody.
  • such anti-CD47 antibody is an IgGl isotype antibody.
  • such anti-CD47 antibody is an IgGl Z allotype isotype antibody.
  • such anti-CD47 antibody is an IgG4, such as an IgG4P or IgG4PE, isotype antibody.
  • Anti-CD47 antibody AB6.12 heavy chain variable region (VH) (Kabat CDRs 1-3 are underlined, SEQ ID NOs: 14-16):
  • Anti-CD47 antibody AB6.12 light chain variable region (VL) (Kabat CDRs 1-3 are underlined, SEQ ID NOs: 17-19):
  • an anti-CD47 described herein comprises one or more amino acid modifications (e.g., 1-15 amino acid modifications), for example in the VH framework region, of a parental antibody, e.g., a parental antibody selected from anti-CD47 antibodies described in U.S. Application Publication No.
  • US 2014/0140989 Al which is hereby incorporated by reference in its entirety, for example anti-CD47 antibodies described in Table 1 of U.S. Application Publication No. US 2014/0140989 Al (e.g., anti-CD47 antibody 2A1, AB2.03, AB2.04, AB2.05, AB2.06, AB2.07, AB2.08, AB2.09, AB2.13, AB3.09, AB6.12, AB6.13, AB6.14, AB6.17, AB10.13, AB10.14, AB11.05, AB12.05, AB15.05, AB16.05, AB17.05, AB22.05, AB23.05, AB24.05, and AB25.05).
  • an antibody e.g. a monoclonal antibody, which specifically binds to human CD47, wherein such an anti-CD47 antibody is a variant of a parental anti-CD47 antibody, wherein the anti-CD47 antibody, when produced using a cell-free (CF) expression system, has a higher antibody expression titer or yield compared to that of the parental anti-CD47 antibody when expressed in the CF system, and wherein the anti-CD47 antibody comprises a VH comprising the following N-terminal to C-terminal sequence:
  • Xi4 ⁇ are ordered from N-terminus to C-terminus, wherein Xi is M or there is no amino acid at position Xi, X 2 is an amino acid with hydrophobic side chains such as M or V, X 3 is T or P, X 4 is S or A, X 5 is an acid having aliphatic side chains such as A or G, X 6 is F or L, X 7 is D or G, X 8 is an amino acid with hydrophobic side chains such as I or M, X 9 is R or T, ⁇ ⁇ is R or T, X u is M or T, X 12 is S or R, X 13 is a negatively charged amino acid such as E or D, and X 14 is an amino acid with hydrophobic side chains such as M or V.
  • an anti-CD47 antibody described herein comprises a VH comprising the sequence of SEQ ID NO: 20, wherein the amino acid at position X 1 is any amino acid such as M, X 2 is not M, X 3 is not T, X 4 is not S, X 5 is not A, X 6 is not F, X 7 is not D, X 8 is not I, Xg is not R, X 10 is not R, Xn is not M, X 12 is not S, X 13 is not E, and/or X 1 is not M.
  • any 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 of Xi to X 14 are not these amino acids.
  • the VH amino acid sequence is not the VH amino acid sequence of antibody AB6.12, for example, the VH amino acid sequence is not SEQ ID NO: 1.
  • an anti-CD47 antibody described herein comprises a VH comprising the sequence of SEQ ID NO: 20, wherein the amino acid at position X 7 is not G X 9 is not A and/or Xn is not S.
  • any 1, 2, or 3 of X 7; X 9 and Xn are not these amino acids.
  • an anti-CD47 antibody described herein comprises a VH comprising the sequence of SEQ ID NO: 20, wherein the amino acid at position X 7 is not G X 8 is not M, X 9 is not E, Xi 0 is not T, and/or Xn is not T. In particular aspects, any 1, 2, 3, or 4 of X 7 to Xn are not these amino acids. In particular aspects, when the amino acid at position X 7 is G then X 8 is M, Xi 0 is T, X 9 is not E, and Xn is T.
  • an anti-CD47 antibody described herein comprises a VH comprising the sequence of SEQ ID NO: 20, wherein the VH does not comprise the amino acid sequence of SEQ ID NO: 5, 6, 7, 8, 9, 10, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, of U.S. Application Publication No. US2014/0140989 Al, which is
  • an anti-CD47 antibody described herein comprises a VH comprising the consensus sequence of SEQ ID NO: 20, wherein the VH does not comprise the framework regions of the amino acid sequence of SEQ ID NO: 5, 6, 7, 8, 9, 10, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, of U.S. Application Publication No. US2014/0140989 Al, which is incorporated herein by reference in its entirety.
  • Xi is M
  • X 2 is V
  • X 3 is P
  • X 4 is A
  • X 5 is G
  • X 6 is L
  • X 7 is G
  • X 8 is
  • Xi is M
  • X 2 is M
  • X 3 is P
  • X 4 is S
  • X 5 is A
  • X 6 is F
  • X 7 is G
  • X 8 is
  • an anti-CD47 antibody provided herein is not antibody AB6.12.
  • an anti-CD47 antibody provided herein does not comprise a VH (e.g., SEQ
  • VL e.g., SEQ ID NO: 12
  • an anti-CD47 antibody provided herein, comprises one of the following VH amino acid sequences presented in Table 1. [0079] Table 1: VH amino acid sequence
  • an antibody e.g. a monoclonal antibody, which specifically binds to human CD47, wherein such an anti-CD47 antibody is a variant of a parental anti-CD47 antibody, wherein the anti-CD47 antibody, when produced using a cell-free (CF) expression system, has a higher antibody expression titer or yield compared to that of the parental anti-CD47 antibody when expressed in the CF system, and wherein the anti-CD47 antibody comprises a VH comprising SEQ ID NO: 21.
  • such anti-CD47 antibody is an IgGl, IgG2, IgG3, or IgG4 isotype antibody.
  • such anti-CD47 antibody is an IgGl isotype antibody. In certain aspects, such anti-CD47 antibody is an IgGl Z allotype isotype antibody. In certain aspects, such anti-CD47 antibody is an IgG4, such as an IgG4P or IgG4PE, isotype antibody.
  • an antibody e.g. a monoclonal antibody, which specifically binds to human CD47, wherein such an anti-CD47 antibody is a variant of a parental anti-CD47 antibody, wherein the anti-CD47 antibody, when produced using a cell-free (CF) expression system, has a higher antibody expression titer or yield compared to that of the parental anti-CD47 antibody when expressed in the CF system, and wherein the anti-CD47 antibody comprises a VH comprising SEQ ID NO: 22.
  • such anti-CD47 antibody is an IgGl, IgG2, IgG3, or IgG4 isotype antibody.
  • such anti-CD47 antibody is an IgGl isotype antibody. In certain aspects, such anti-CD47 antibody is an IgGl Z allotype isotype antibody. In certain aspects, such anti-CD47 antibody is an IgG4, such as an IgG4P or IgG4PE, isotype antibody.
  • an anti-CD47 antibody (IgGl -13m) provided herein comprises an IgGl heavy chain comprising the amino acid sequence as set forth below: MQVQLVQSGAEVKKPGASVKVSCKASGFNIKDYYLHWVRQAPGQGLEWMGWIDPDQGDTEYAQK LQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCNAAYGSSSYPMDYWGQGTTVTVSSASTKGPS VFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSWTV PSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLM ISRTPEVTCWVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRWSVLTVLHQDWLNG KEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTL
  • an anti-CD47 antibody (IgGl-13mZ ) provided herein comprises an IgGl-Z allotype heavy chain comprising the amino acid sequence as set forth below:
  • an anti-CD47 antibody (IgGl-5m) provided herein comprises an IgGl heavy chain comprising the amino acid sequence as set forth below:
  • an anti-CD47 antibody (IgG4P-13m) provided herein comprises an IgG4P antibody comprising the amino acid sequence as set forth below: MQVQLVQSGAEVKKPGASVKVSCKASGFNIKDYYLHWVRQAPGQGLEWMGWIDPDQGDTEYAQK LQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCNAAYGSSSYPMDYWGQGTTVTVSSASTKGPS VFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSWTV PSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFLGGPSVFLFPPKPKDTLMISR TPEVTCVWDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRWSVLTVLHQDWLNGKEY KCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPS
  • an anti-CD47 antibody (IgG4P-5m) provided herein comprises an IgG4P heavy chain comprising the amino acid sequence as set forth below:
  • an anti-CD47 antibody (IgG4PE-13m) provided herein comprises an IgG4PE heavy chain comprising the amino acid sequence as set forth below:
  • an anti-CD47 antibody (IgG4PE-5m) provided herein comprises an IgG4PE heavy chain comprising the amino acid sequence as set forth below:
  • an antibody e.g. a monoclonal antibody, which specifically binds to human CD47, wherein such an anti-CD47 antibody is a variant of a parental anti-CD47 antibody, wherein the anti-CD47 antibody, when produced using a cell-free (CF) expression system, has a higher antibody expression titer or yield compared to that of the parental anti-CD47 antibody when expressed in the CF system, and wherein the anti-CD47 antibody comprises a light chain comprising a kappa or lambda light chain constant region (e.g., human kappa or lambda light chain constant region), for example SEQ ID NO: 13.
  • CF cell-free
  • an antibody e.g. a monoclonal antibody, which specifically binds to human CD47, wherein such an anti-CD47 antibody is a variant of a parental anti-CD47 antibody, wherein the anti-CD47 antibody, when produced using a cell-free (CF) expression system, has a higher antibody expression titer or yield compared to that of the parental anti-CD47 antibody when expressed in the CF system, and wherein the anti-CD47 antibody comprises (i) a VH described herein (e.g., SEQ ID NO: 20, 21, or 22) or a heavy chain described herein (e.g., any one of SEQ ID NOs:5-l 1), and (ii) a light chain comprising a kappa or lambda light chain constant region (e.g., human kappa or lambda light chain constant region), for example SEQ ID NO: 13, e.g., as set forth below (anti-CD47 antibody light chain (I
  • an anti-CD47 described herein is not an anti-CD47 antibody described in U.S. Application Publication No. US 2014/0140989 Al, which is hereby incorporated by reference in its entirety, for example any one of anti-CD47 antibodies in Table 1 of the publication (e.g., anti-CD47 antibody 2A1, AB2.03, AB2.04, AB2.05, AB2.06, AB2.07, AB2.08, AB2.09, AB2.13, AB3.09, AB6.12, AB6.13, AB6.14, AB6.17, AB10.13, AB10.14, AB11.05, AB 12.05, AB 15.05, AB16.05, AB17.05, AB22.05, AB23.05, AB24.05, and AB25.05), or any antibody comprising any of SEQ ID NOS: 5-30 of the publication.
  • an anti-CD47 antibody provided herein or an antigen-binding fragment thereof is an IgG isotype.
  • the constant region of the antibody is of human IgGl isotype, having an amino acid sequence:
  • the human IgGl constant region is modified at amino acid Asn297 (Boxed, Kabat Numbering) to prevent to glycosylation of the antibody, for example Asn297Ala (N297A).
  • the constant region of the antibody is modified at amino acid Leu235 (Kabat Numbering) to alter Fc receptor interactions, for example Leu235Glu (L235E) or Leu235Ala (L235A).
  • the constant region of the antibody is modified at amino acid Leu234 (Kabat Numbering) to alter Fc receptor interactions, e.g.,
  • Leu234Ala (L234A).
  • the constant region of the antibody is altered at both amino acid 234 and 235, for example Leu234Ala and Leu235Ala (L234A/L235A) (EU index of Kabat et al 1991 Sequences of Proteins of Immunological Interest).
  • the constant region of an anti-CD47 antibody provided herein is of human IgG2 isotype, having an amino acid sequence:
  • the human IgG2 constant region is modified at amino acid
  • Asn297 (Boxed, Kabat Numbering) to prevent to glycosylation of the antibody, e.g., Asn297Ala (N297A).
  • the constant region of an anti-CD47 antibody provided herein is of human IgG3 isotype, having an amino acid sequence:
  • the human IgG3 constant region is modified at amino acid
  • the human IgG3 constant region is modified at amino acid 435 to extend the half-life, e.g., Arg435His (R435H) (EU index of Kabat et al 1991 Sequences of Proteins of Immunological Interest).
  • the constant region of an anti-CD47 antibody provided herein is of human IgG4 isotype, having an amino acid sequence:
  • the human IgG4 constant region is modified within the hinge region to prevent or reduce strand exchange, e.g., Ser228Pro (S228P).
  • the human IgG4 constant region is modified at amino acid 235 to alter Fc receptor interactions, e.g., Leu235Glu (L235E).
  • the human IgG4 constant region is modified within the hinge and at amino acid 235, e.g., Ser228Pro and Leu235Glu (S228P/L235E).
  • the human IgG4 constant region is modified at amino acid Asn297 (Kabat Numbering) to prevent to glycosylation of the antibody, e.g., Asn297Ala (N297A).
  • the human IgG4 constant region is modified at amino acid positions Ser228, Leu235, and Asn297 (e.g., S228P/L235E/N297A). (EU index of Kabat et al 1991 Sequences of Proteins of Immunological Interest).
  • the antibody is of human IgG4 subclass and lacks glycosylation.
  • the glycosylation can be eliminated by mutation at position 297 (Kabat numbering), for example N297A.
  • the glycosylation can be eliminated by production of the antibody in a host cell that lacks the ability for post-translational glycosylation, for example a bacterial or yeast derived system or a modified mammalian cell expression system.
  • the human IgG constant region is modified to alter antibody-dependent cellular cytotoxicity (ADCC) and/or complement-dependent cytotoxicity (CDC), e.g., the amino acid modifications described in Natsume et al., 2008 Cancer Res, 68(10): 3863-72; Idusogie et al., 2001 J Immunol, 166(4): 2571-5; Moore et al., 2010 mAbs, 2(2): 181- 189; Lazar et al., 2006 PNAS, 103(11): 4005-4010, Shields et al., 2001 JBC, 276( 9): 6591- 6604; Stavenhagen et al., 2007 Cancer Res, 67(18): 8882-8890; Stavenhagen et al., 2008 Advan. Enzyme Regul., 48: 152-164; Alegre et al, 1992 J Immunol, 148: 3461-3468; Reviewed in Kan
  • ADCC antibody-dependent
  • the human IgG constant region is modified to induce heterodimerization.
  • having an amino acid modification within the CH3 domain at Thr366, which when replaced with a more bulky amino acid, e.g., Try (T366W) is able to preferentially pair with a second CH3 domain having amino acid modifications to less bulky amino acids at positions Thr366, Leu368, and Tyr407, e.g., Ser, Ala and Val, respectively (T366S/L368A/Y407V).
  • Heterodimerization via CH3 modifications can be further stabilized by the introduction of a disulfide bond, for example by changing Ser354 to Cys (S354C) and Y349 to Cys (Y349C) on opposite CH3 domains (Reviewed in Carter, 2001 Journal of Immunological Methods, 248: 7-15).
  • the antibody lacks glycosylation, but is not modified at amino acid Asn297 (Kabat numbering).
  • the glycosylation can, for example, be eliminated by production of the antibody in a host cell that lacks a post-translational glycosylation capacity, for example a bacterial or yeast derived system or a modified mammalian cell expression system.
  • a host cell that lacks a post-translational glycosylation capacity, for example a bacterial or yeast derived system or a modified mammalian cell expression system.
  • such a system can be a CF expression system.
  • an anti-CD47 antibody described herein or an antigen- binding fragment thereof comprises amino acid sequences with certain percent identity relative to a parental antibody.
  • the determination of percent identity between two sequences can be accomplished using a mathematical algorithm.
  • a non-limiting example of a mathematical algorithm utilized for the comparison of two sequences is the algorithm of Karlin and Altschul, 1990, Proc. Natl. Acad. Sci. U.S.A. 87:2264 2268, modified as in Karlin and Altschul, 1993, Proc. Natl. Acad. Sci. U.S.A. 90:5873 5877.
  • Such an algorithm is incorporated into the NBLAST and XBLAST programs of Altschul et al., 1990, J. Mol. Biol. 215:403.
  • Gapped BLAST can be utilized as described in Altschul et al, 1997, Nucleic Acids Res. 25:3389 3402.
  • PSI BLAST can be used to perform an iterated search which detects distant relationships between molecules (Id.).
  • the default parameters of the respective programs e.g., of XBLAST and NBLAST
  • the default parameters of the respective programs e.g., of XBLAST and NBLAST
  • the default parameters of the respective programs e.g., of XBLAST and NBL
  • NCBI Biotechnology Information
  • Another preferred, non limiting example of a mathematical algorithm utilized for the comparison of sequences is the algorithm of Myers and Miller, 1988, CABIOS 4: 11 17.
  • Such an algorithm is incorporated in the ALIGN program (version 2.0) which is part of the GCG sequence alignment software package.
  • ALIGN program version 2.0
  • an antibody described herein or an antigen-binding fragment thereof comprises a VL domain having at least 80%, at least 85%>, at least 90%, at least 95%, at least 98%), or at least 99% sequence identity to the amino acid sequence of SEQ ID NO: 12, wherein the antibody specifically binds to CD47.
  • an antibody described herein or an antigen-binding fragment thereof comprises a VL domain having at least 80%>, at least 85%), at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO: 12, wherein the antibody specifically binds to CD47, and wherein the antibody comprises CDRs (e.g., VL CDRs 1-3) that are identical to the CDRs (e.g., VL CDRs 1-3) of SEQ ID NO: 12 (e.g., SEQ ID NO: 17-19).
  • CDRs e.g., VL CDRs 1-3
  • an antibody described herein or an antigen-binding fragment thereof comprises a light chain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%), or at least 99% sequence identity to the amino acid sequence of SEQ ID NO: 13, wherein the antibody specifically binds to CD47.
  • an antibody described herein or an antigen-binding fragment thereof comprises a light domain having at least 80%, at least 85%), at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO: 13, wherein the antibody specifically binds to CD47, and wherein the antibody comprises CDRs (e.g., VL CDRs 1-3) that are identical to the CDRs (e.g., VL CDRs 1-3) of SEQ ID NO: 13 (e.g., SEQ ID NO: 17-19).
  • CDRs e.g., VL CDRs 1-3
  • an antibody described herein or an antigen-binding fragment thereof comprises a VH domain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%), or at least 99% sequence identity to the amino acid sequence of SEQ ID NO: 1, wherein the antibody specifically binds to CD47 and wherein the anti-CD47 antibody, when produced using a cell-free expression system, has a higher antibody expression titer or yield compared to the parental antibody when produced in the CF expression system.
  • an antibody described herein or an antigen-binding fragment thereof comprises a VH domain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99%) sequence identity to the amino acid sequence of SEQ ID NO: 1, wherein the antibody specifically binds to CD47, and wherein the antibody comprises CDRs (e.g., VL CDRs 1-3) that are identical to the CDRs (e.g., VL CDRs 1-3) of SEQ ID NO: 1 (e.g., SEQ ID NO: 14-16).
  • CDRs e.g., VL CDRs 1-3
  • an antibody described herein or an antigen-binding fragment thereof comprises a light chain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO: 2, wherein the antibody specifically binds to CD47 and wherein the anti-CD47 antibody, when produced using a cell-free expression system, has a higher antibody expression titer or yield compared to the parental antibody when produced in the CF expression system.
  • an antibody described herein or an antigen-binding fragment thereof comprises a heavy domain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO: 2, wherein the antibody specifically binds to CD47, and wherein the antibody comprises CDRs (e.g., VL CDRs 1-3) that are identical to the CDRs (e.g., VL CDRs 1-3) of SEQ ID NO: 2 (e.g., SEQ ID NO: 17-19).
  • CDRs e.g., VL CDRs 1-3
  • an antibody described herein or an antigen-binding fragment thereof comprises a light chain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%), or at least 99% sequence identity to the amino acid sequence of SEQ ID NO: 3, wherein the antibody specifically binds to CD47 and wherein the anti-CD47 antibody, when produced using a cell-free expression system, has a higher antibody expression titer or yield compared to the parental antibody when produced in the CF expression system.
  • an antibody described herein or an antigen-binding fragment thereof comprises a heavy domain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO: 3, wherein the antibody specifically binds to CD47, and wherein the antibody comprises CDRs (e.g., VL CDRs 1-3) that are identical to the CDRs (e.g., VL CDRs 1-3) of SEQ ID NO: 3 (e.g., SEQ ID NO: 17-19).
  • CDRs e.g., VL CDRs 1-3
  • an antibody described herein or an antigen-binding fragment thereof comprises a light chain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%), or at least 99% sequence identity to the amino acid sequence of SEQ ID NO: 4, wherein the antibody specifically binds to CD47 and wherein the anti-CD47 antibody, when produced using a cell-free expression system, has a higher antibody expression titer or yield compared to the parental antibody when produced in the CF expression system.
  • an antibody described herein or an antigen-binding fragment thereof comprises a heavy domain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99%) sequence identity to the amino acid sequence of SEQ ID NO: 4, wherein the antibody specifically binds to CD47, and wherein the antibody comprises CDRs (e.g., VL CDRs 1-3) that are identical to the CDRs (e.g., VL CDRs 1-3) of SEQ ID NO: 4 (e.g., SEQ ID NO: 17-19).
  • CDRs e.g., VL CDRs 1-3
  • anti-CD47 antibodies provided herein exhibit one or more desirable characteristics, such as, by way of non-limiting example, blocking of the interaction between CD47 and its ligand SIRPa and/or promoting (e.g., inducing or increasing)
  • anti-CD47 antibodies provided herein block at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 95%, or at least 99% of the interaction between CD47 and SIRPa as compared to the level of interaction between CD47 and SIRPa in the absence of the anti-CD47 antibody described herein.
  • anti-CD47 antibodies described herein promote (e..g, induce or increase) phagocytosis of cells, e.g., CD47-expressing cells (e.g., CCRF-CEM cells), for example, by macrophages.
  • the level of phagocytosois in the presence of anti- CD47 antibodies described herein is increased by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 99%), at least 150%, at least 200%, compared to the level of aphagocytosis in the presence of anti-CD47 antibodies described herein.
  • anti-CD47 antibodies described herein do not promote (e..g, induce or increase), or cause a significant level of, agglutination of cells, e.g., anti-CD47 antibodies described herein do not promote (e..g, induce or increase), or cause a significant level of, hemagglutination of red blood cells.
  • the level of agglutination in the presence of anti-CD47 antibodies described herein is reduced by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 99% compared to the level of agglutination in the presence of anti-CD47 antibodies known to induce agglutination, such as MCA91 1 mouse anti-human CD47 antibody (BRIC 126).
  • anti-CD47 antibodies described herein do not promote (e.g., induce or increase), or cause a significant level of, agglutination if the level of agglutination in the presence of anti- CD47 antibodies described herein is reduced by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 99% compared to the level of agglutination in the presence of existing anti-CD47 antibodies known to induce agglutination, such as MCA91 1 mouse anti-human CD47 antibody (BRIC126).
  • BRIC126 mouse anti-human CD47 antibody
  • Anti-CD47 antibodies described herien also include monoclonal antibodies that specifically bind CD47, wherein the antibody does not promote (e.g., induce or increase), or cause a significant level of, agglutination, e.g., red blood cell hemagglutination ("RBC
  • the level of RBC depletion is determined by measuring the RBC count in a subject after administration of a treatment, e.g., an anti-CD47 antibody described herein.
  • anti-CD47 antibodies described herein do not promote (e.g., induce or increase), or cause a significant level of, RBC depletion if the RBC count in a subject after administration of an anti-CD47 antibody described herein is within the range of a normal, healthy subject.
  • the RBC count for a normal, healthy male human is about 4.7 to about 6.1 million cells per microliter of blood sample.
  • the RBC count for a normal, healthy female human is 4.2 to about 5.4 million cells per microliter of blood sample.
  • anti-CD47 antibodies described herein do not promote (e.g., induce or increase), or cause a significant level of, RBC depletion if the RBC count in a subject after administration (e.g., 5 min, 10 min, 30 min, 1 h, 2 h, 3 h, 4 h, 5 h, 12 h, 24 h, 2 days, 4 days, 6 days, 1 week, 2 weeks, 3 weeks, 1 month, 2 months, or more) of an anti-CD47 antibody described herein is at least 50%, 60%, 70%, 80%, 90%, 95%, 97%, 98%, 99%, or 99.5% of the RBC count prior to administration.
  • RBC count in a subject after administration e.g., 5 min, 10 min, 30 min, 1 h, 2 h, 3 h, 4 h, 5 h, 12 h, 24 h, 2 days, 4 days, 6 days, 1 week, 2 weeks, 3 weeks, 1 month, 2 months, or more
  • anti-CD47 antibodies described herein do not promote (e.g., induce or increase), or cause a significant level of, RBC depletion if the RBC count in a subject after administration (5 min, 10 min, 30 min, 1 h, 2 h, 3 h, 4 h, 5 h, 12 h, 24 h, 2 days, 4 days, 6 days, 1 week, 2 weeks, 3 weeks, 1 month, 2 months, or more) of an anti47 antibody described herein is at least 50%, 60%, 70%, 80%, 90%, 95%, 97%, 98%, 99%, or 99.5% of the RBC count in a subject after administration of a placebo treatment (e.g., vehicle).
  • RBC counts are determined by standard methods in the art.
  • anti-CD47 antibodies described herein do not promote (e.g., induce or increase), or cause a significant level of, platelet depletion.
  • induce or increase e.g., induce or increase
  • administering leads to a percentage of platelets remaining of at least 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100%.
  • anti-CD47 antibodies described herien include but are not limited to antibodies that do not bind to, or have a low binding affinity to, a Fey receptor (FcyR).
  • FcyR Fey receptor
  • the constant region of an anti-CD47 antibody e.g., when produced using a CF expression system, has a lower binding affinity to a FcyR than the constant region of an anti-CD47 antibody, e.g., when produced using a host cell (e.g., CHO cells) expression system.
  • the level of agglutination e.g., the level of hemagglutination of RBCs.
  • the level of hemagglutination is ascertained by measuring the area of an RBC dot after performing a hemagglutination assay in the presence of anti-CD47 antibodies described, as described in the Examples below. In some cases, the area of the RBC dot in the presence of anti-CD47 antibody described herien is compared to the area of the RBC dot in the absence of an anti-CD47 antibody, e.g., in the presence of zero
  • hemagglutination is quantified relative to a baseline control.
  • a larger RBC dot area corresponds to a higher level of hemagglutination.
  • densitometry of the RBC dot may also be utilized to quantitate hemagglutination.
  • the level of RBC depletion is ascertained, e.g., by measuring the RBC count (i.e., the total number of RBCs in a sample of blood), e.g., by using a cell counter or a hemacytometer.
  • the RBCs in a sample of blood can optionally be isolated by fractionating whole blood using, e.g., centrifugation, prior to counting.
  • the RBC count in the presence of an anti-CD47 antibody described herein is compared to the RBC count in the absence of the CD47 antibody, e.g., in the presence of zero RBC depletion. In this manner, the level of RBC depletion is normalized relative to a baseline control.
  • anti-CD47 antibodies provided herein exhibit inhibitory activity, for example by inhibiting CD47 expression (e.g., inhibiting cell surface expression of CD47), activity, and/or signaling, or by interfering with the interaction between CD47 and SIRPa.
  • anti-CD47 antibodies provided herein completely or partially reduce or otherwise modulate CD47 expression or activity upon binding to, or otherwise interacting with, CD47, e.g., a human CD47. The reduction or modulation of a biological function of CD47 is complete, significant, or partial upon interaction between the antibodies and the human CD47 polypeptide and/or peptide.
  • Anti-CD47 antibodies described herein are considered to completely inhibit CD47 expression or activity when the level of CD47 expression or activity in the presence of the antibody is decreased by at least 95%, e.g., by 96%, 97%, 98%, 99% or 100% as compared to the level of CD47 expression or activity in the absence of interaction, e.g., binding, with the antibody described herein.
  • anti-CD47 antibodies are considered to significantly inhibit CD47 expression or activity when the level of CD47 expression or activity in the presence of the CD47 antibody is decreased by at least 50%, e.g., 55%, 60%, 75%, 80%, 85%) or 90% as compared to the level of CD47 expression or activity in the absence of binding with a CD47 antibody described herein.
  • anti-CD47 antibodies are considered to partially inhibit CD47 expression or activity when the level of CD47 expression or activity in the presence of the antibody is decreased by less than 95%, e.g., 10%, 20%, 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85% or 90% as compared to the level of CD47 expression or activity in the absence of interaction, e.g., binding, with an antibody described herein.
  • anti-CD47 antibodies provided herein comprise one or more non-natural amino acid residues at site-specific positions. See, e.g., U.S. Application Publication No. US 2014/0046030 Al, which is incorporated herein by reference in its entirety.
  • non-natural amino acid residues at site specific positions has advantages for antibody production yield, solubility, binding affinity, and/or activity.
  • Non-limiting examples of non- natural amino acids have been described, see, e.g., U.S. Application Publication No. US
  • a conjugation moiety can be any conjugation moiety deemed useful to one of skill in the art.
  • a conjugation moiety can be a polymer, such as polyethylene glycol, that can improve the stability of the antibody in vitro or in vivo.
  • a conjugation moiety can have therapeutic activity, thereby yielding an antibody-drug conjugate.
  • a conjugation moiety can be a molecular payload that is harmful to target cells.
  • a conjugation moiety can be a label useful for detection or diagnosis.
  • a conjugation moiety is linked to the antibody via a direct covalent bond.
  • a conjugation moiety is linked to the antibody via a linker.
  • a conjugation moiety or a linker is attached via one of the non-natural amino acids of an anti-CD47 antibody.
  • Exemplary conjugation moieties and linkers have been described, e.g., see U.S.
  • Antibodies or an antigen-binding fragments described herein that immunospecifically bind to CD47 can be produced by any method known in the art, for example, by chemical synthesis or by recombinant expression techniques (e.g., CF expression systems).
  • Such methods can employ conventional techniques in molecular biology, microbiology, genetic analysis, recombinant DNA, organic chemistry, biochemistry, PCR, oligonucleotide synthesis and modification, nucleic acid hybridization, and related fields within the skill of the art. These techniques are described, for example, in the references cited herein and are fully explained in the literature. See, e.g.,, Maniatis et al. (1982) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press; Sambrook et al. (1989), Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor Laboratory Press;
  • anti-CD47 antibodies as provided herein can be produced using a CF expression system, for example, a CF expression system as known in the art, and, for example, as described in the Examples below.
  • CF expression systems can include cell-free extracts, such as S30 cell-free extracts, with DsbC, and 20 amino acids (e.g., natural or non-natural), and optionally, one or more of iodoacetamide, magnesium glutamate, ammonium glutamate, mM potassium glutamate, sodium pyruvate, AMP, GMP, UMP, and CMP, sodium oxalate, putrescine, spermidine, potassium phosphate, T7 RNAP, and oxidized (GSSG) glutathione.
  • Heavy chain plasmids and light chain plasmids are added accordingly to the CF extract composition for polypeptide production and purification.
  • the CF expression system is an in vitro transcription and translation system as described in Yin et al., mAbs, 2012, 4:217-225, incorporated by reference in its entirety.
  • the cell-free system utilizes a cell-free extract from a eukaryoctic cell or from a prokaryotic cell.
  • the prokaryotic cell is E. coli.
  • the CF expression system can utilize a system as described in US Application Publication No. US 2014/0315245, which is hereby incorporated by reference in its entirety.
  • the CF expression system can comprise a bacterial extract having an oxidative phosphorylation system and components necessary for cell free protein synthesis and, in certain embodiments, can further comprise an exogenous protein chaperone, e.g., a protein disulfide isomerase (PDI), or a peptide-prolyl cis-trans isomerase.
  • PDI protein disulfide isomerase
  • the PDI is a member of the Dsb (disulfide bond formation) family of E. coli, for example, DsbA or DsbC.
  • the CF expression system comprises a cell extract of £ coli strain SBDG028, SBDG031, or SBDG044, as described in US Application Publication No. US 2014/0315245, which can, for example, be prepared according to Zawada et al., Biotechnology and Bioengineering (2011) vol. 108, No. 7.
  • anti-CD47 antibodies provided herein comprise amino acid modifications that allow for antibody production using CF expression systems better than the parental antibodies.
  • anti-CD47 antibodies provided herein which are produced using CF expression systems are aglycosylated.
  • Monoclonal antibodies can, for example, be prepared using a wide variety of techniques known in the art including the use of hybridoma, recombinant, and phage display technologies, or a combination thereof.
  • monoclonal antibodies can be produced using hybridoma techniques including those known in the art and taught, for example, in Harlow et al, Antibodies: A Laboratory Manual (Cold Spring Harbor Laboratory Press, 2nd ed. 1988); Hammerling et al, in: Monoclonal Antibodies and T-Cell Hybridomas 563 681 (Elsevier, N.Y., 1981).
  • the term "monoclonal antibody” as used herein is not limited to antibodies produced through hybridoma technology.
  • monoclonal antibodies can be produced recombinantly from host cells engineered to express an antibody described herein (e.g., anti- CD47 antibody comprising the CDRs of any one of antibodies Ab235-Ab255) or a fragment thereof, for example, a light chain and/or heavy chain of such an antibody.
  • an antibody described herein e.g., anti- CD47 antibody comprising the CDRs of any one of antibodies Ab235-Ab255
  • a fragment thereof for example, a light chain and/or heavy chain of such an antibody.
  • the antibodies described herein or antigen-binding fragments thereof can also be generated using various phage display methods known in the art.
  • phage display methods functional antibody domains are displayed on the surface of phage particles which carry the polynucleotide sequences encoding them.
  • Examples of phage display methods that can be used to make the antibodies described herein include those disclosed in Brinkman et al, 1995, J. Immunol. Methods 182:41-50; Ames et al, 1995, J. Immunol. Methods 184: 177-186;
  • Antibodies described herein can, for example, include chimeric antibodies.
  • a chimeric antibody is a molecule in which different portions of the antibody are derived from different immunoglobulin molecules.
  • a chimeric antibody can contain a variable region of a mouse or rat monoclonal antibody fused to a constant region of a human antibody.
  • Methods for producing chimeric antibodies are known in the art. See, e.g., Morrison, 1985, Science 229: 1202; Oi et al, 1986, BioTechniques 4:214; Gillies et al, 1989, J. Immunol.
  • Antibodies or antigen-binding fragments produced using techniques such as those described herein can be isolated using standard, well known techniques.
  • antibodies or antigen-binding fragments can be suitably separated from, e.g., culture medium, ascites fluid, serum, cell lysate, synthesis reaction material or the like by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose,_hydroxylapatite
  • an "isolated” or “purified” antibody is substantially free of cellular material or other proteins from the cell or tissue source from which the antibody is derived, or substantially free of chemical precursors or other chemicals when chemically synthesized, or from the components of the CF expression system used to produce the antibodies.
  • Antibodies described herein include antibody fragments which recognize specific CD47 antigens and can be generated by any technique known to those of skill in the art.
  • Fab and F(ab'> 2 fragments described herein can be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab' )2 fragments).
  • a Fab fragment corresponds to one of the two identical arms of an antibody molecule and contains the complete light chain paired with the VH and CHI domains of the heavy chain.
  • a F(ab' )2 fragment contains the two antigen-binding arms of an antibody molecule linked by disulfide bonds in the hinge region.
  • antibody fragments described herein can routinely be produced via well known recombinant expression techniques. See, e.g., PCT publication No. WO 92/22324; Mullinax et al, 1992, BioTechniques 12(6):864-869; Sawai et al, 1995, AJRI 34:26-34; and Better et al, 1988, Science 240: 1041 - 1043.
  • Antibodies described herein can, for example, include humanized antibodies, e.g., deimmunized or composite human antibodies.
  • a humanized antibody can comprise human constant region sequences.
  • a humanized antibody can be selected from any class of immunoglobulins, including IgM, IgG, IgD, IgA and IgE, and any isotype, including IgGi, IgG 2 , IgG 3 and IgG 4 .
  • a humanized antibody can comprise kappa or lambda light chain constant sequences.
  • Humanized antibodies can be produced using a variety of techniques known in the art, including but not limited to, CDR-grafting (European Patent No. EP 239,400; International publication No. WO 91/09967; and U.S. Patent Nos. 5,225,539, 5,530, 101, and 5,585,089), veneering or resurfacing (European Patent Nos. EP 592, 106 and EP 519,596; Padlan, 1991, Molecular Immunology 28(4/5):489-498; Studnicka et al, 1994, Protein Engineering 7(6):805- 814; and Roguska et a/., 1994, PNAS 91 :969-973), chain shuffling (U.S. Patent No.
  • Antibodies described herein can, for example, be multispecific, e.g., bispecific, antibodies. Methods for making multispecific (e.g, bispecific antibodies) have been described, see, for example, U.S. Patent Nos. 7951917, 7183076, 8227577, 5837242, 5989830, 5869620, 6132992, and 8586713.
  • Single domain antibodies for example, antibodies lacking the light chains, can be produced by methods well-known in the art. See Riechmann et al., 1999, J. Immunol. 231 :25-38; Nuttall et al, 2000, Curr. Pharm. Biotechnol.
  • Human antibodies can be produced using any method known in the art. For example, well known transgenic mice which are incapable of expressing functional endogenous murine immunoglobulins, but which can express human immunoglobulin genes, can be used.
  • phage display techniques described above, can be utilized.
  • human antibodies can, for example, be produced using mouse- human hybridomas.
  • human peripheral blood lymphocytes transformed with Epstein-Barr virus (EBV) can be fused with mouse myeloma cells to produce mouse-human hybridomas secreting human monoclonal antibodies, and these mouse-human hybridomas can be screened to determine ones which secrete human monoclonal antibodies that
  • Antibody variable domains with the desired binding specificities can be fused to immunoglobulin constant domain sequences.
  • the fusion preferably is with an immunoglobulin heavy-chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is preferred to have the first heavy-chain constant region (CHI) containing the site necessary for light-chain binding present in at least one of the fusions.
  • CHI first heavy-chain constant region
  • the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers which are recovered from recombinant cell culture.
  • the preferred interface comprises at least a part of the CH3 region of an antibody constant domain.
  • one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g. tyrosine or tryptophan).
  • Compensatory "cavities" of identical or similar size to the large side chain(s) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine). This provides a mechanism for increasing the yield of the heterodimer over other unwanted end-products such as
  • Bispecific antibodies can be prepared as full length antibodies or antibody fragments (e.g. F(ab') 2 bispecific antibodies). Techniques for generating bispecific antibodies from antibody fragments have been described in the literature. For example, bispecific antibodies can be prepared using chemical linkage. Brennan et al., Science 229:81 (1985) describe a procedure wherein intact antibodies are proteolytically cleaved to generate F(ab') 2 fragments. These fragments are reduced in the presence of the dithiol complexing agent sodium arsenite to stabilize vicinal dithiols and prevent intermolecular disulfide formation. The Fab' fragments generated are then converted to thionitrobenzoate (T B) derivatives.
  • T B thionitrobenzoate
  • One of the Fab'-T B derivatives is then reconverted to the Fab'-thiol by reduction with mercaptoethylamine and is mixed with an equimolar amount of the other Fab'-TNB derivative to form the bispecific antibody.
  • the bispecific antibodies produced can be used as agents for the selective Fab'-T B derivatives.
  • polynucleotides comprising a nucleotide sequence encoding an antibody described herein or a fragment thereof (e.g., a variable light chain region and/or variable heavy chain region) that immunospecifically binds to a CD47 antigen
  • vectors e.g., vectors comprising such polynucleotides for recombinant expression in host cells (e.g., E. coli and mammalian cells).
  • host cells e.g., E. coli and mammalian cells.
  • cells e.g., host cells.
  • methods of making the antibodies and antigen-binding fragments described herein are also provided herein.
  • polynucleotides comprising a nucleotide sequence encoding the light chain or heavy chain of an antibody described herein.
  • polynucleotides comprising a nucleotide sequence encoding the light chain and heavy chain of an antibody described herein.
  • the polynucleotides can comprise nucleotide sequences encoding a light chain comprising the VL FRs and CDRs of antibodies described herein.
  • the polynucleotides can comprise nucleotide sequences encoding a heavy chain comprising the VH FRs and CDRs of antibodies described herein.
  • a polynucleotide described herein encodes a VL chain region of SEQ ID NO: 13 or SEQ ID NO: 13 without amino acid M at the N-terminus. In specific embodiments, a polynucleotide described herein encodes a VH chain region of any one of SEQ ID NOs: 5-11 20-22. In specific embodiments, a polynucleotide described herein encodes a light chain comprising the amino acid sequence of SEQ ID NO: 13. In specific embodiments, a polynucleotide described herein encodes a heavy chain comprising the amino acid sequence of any one of SEQ ID NOs: 5-11.
  • a polynucleotide described herein comprises a nucleotide sequence provided in Table 2 encoding a light chain or a heavy chain of an anti-CD47 antibody provided here.
  • Anti-CD47 IgGl-13mZ HC
  • a polynucleotide provided herein is operably linked to a promoter for expression of such polynucleotide sequence in a host cell.
  • the promoter is derived form the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter, the vaccinia virus 7.5K promoter).
  • the expression of nucleotide sequences encoding antibodies described herein is regulated by a constitutive promoter, inducible promoter or tissue specific promoter.
  • a polynucleotide comprising a nucleotide sequence encoding an antibody comprising a light chain and a heavy chain, e.g., a separate light chain and heavy chain.
  • a light chain in a specific embodiment, a light chain and a heavy chain.
  • polynucleotide provided herein comprises a nucleotide sequence encoding a kappa light chain.
  • a polynucleotide provided herein comprises a nucleotide sequence encoding a lambda light chain.
  • a polynucleotide provided herein comprises a nucleotide sequence encoding an antibody described herein comprising a human kappa light chain or a human lambda light chain.
  • human constant region sequences can be those described in U.S. Patent No. 5,693,780.
  • a polynucleotide provided herein comprises a nucleotide sequence encoding an antibody described herein, which immunospecifically binds to a CD47 polypeptide, wherein the antibody comprises a heavy chain, wherein the constant region of the heavy chain comprises the amino acid sequence of a human gamma ( ⁇ ) heavy chain constant region, for example, human gamma ( ⁇ ) 1 heavy chain constant region, human gamma ( ⁇ ) 2 heavy chain constant region, human gamma ( ⁇ ) 3 heavy chain constant region, or human gamma ( ⁇ ) 4 heavy chain constant region.
  • a human gamma ( ⁇ ) heavy chain constant region for example, human gamma ( ⁇ ) 1 heavy chain constant region, human gamma ( ⁇ ) 2 heavy chain constant region, human gamma ( ⁇ ) 3 heavy chain constant region, or human gamma ( ⁇ ) 4 heavy chain constant region.
  • cells e.g., host cells
  • cells expressing (e.g., recombinantly) antibodies described herein (or an antigen-binding fragment thereof) which specifically bind to CD47 and related polynucleotides and expression vectors, for example, polynucleotides and expression vectors suitable for use in CF expression systems.
  • vectors e.g., expression vectors
  • methods for producing an anti- CD47 antibody described herein comprising expressing such an antibody using a CF expression system, for example, under conditions resulting in improved antibody expression titer or yield.
  • vectors e.g., expression vectors
  • host cells comprising such vectors for recombinantly expressing anti-CD47 antibodies described herein (e.g., human or humanized antibody).
  • methods for producing an antibody described herein, comprising expressing such an antibody using host cells comprising expressing such an antibody using host cells.
  • Recombinant expression of an antibody described herein e.g., a full-length antibody, heavy and/or light chain of an antibody, or a single chain antibody described herein
  • an antibody described herein e.g., a full-length antibody, heavy and/or light chain of an antibody, or a single chain antibody described herein
  • Recombinant expression of an antibody described herein involves construction of an expression vector containing a
  • polynucleotide that encodes the antibody Once a polynucleotide encoding an antibody molecule, heavy and/or light chain of an antibody, or a fragment thereof (e.g., heavy and/or light chain variable domains) described herein has been obtained, the vector for the production of the antibody molecule can be produced by recombinant DNA technology using techniques well- known in the art. Thus, methods for preparing a protein by expressing a polynucleotide containing an antibody or antibody fragment (e.g., light chain or heavy chain) encoding nucleotide sequence are described herein.
  • Such vectors can, for example, include the nucleotide sequence encoding the constant region of the antibody molecule (see, e.g., International Publication Nos. WO 86/05807 and WO 89/01036; and U.S. Patent No. 5, 122,464) and variable domains of the antibody can be cloned into such a vector for expression of the entire heavy, the entire light chain, or both the entire heavy and light chains.
  • An expression vector can be transferred to a cell (e.g., host cell) by conventional techniques and the resulting cells can then be cultured by conventional techniques to produce an antibody described herein or a fragment thereof.
  • a cell e.g., host cell
  • host cells containing a polynucleotide encoding an antibody described herein or fragments thereof, or a heavy or light chain thereof, or fragment thereof, or a single chain antibody described herein, operably linked to a promoter for expression of such sequences in the host cell.
  • vectors encoding both the heavy and light chains, individually can be co-expressed in the host cell for expression of the entire immunoglobulin molecule, as detailed below.
  • a host cell contains a vector comprising a polynucleotide encoding both the heavy chain and light chain of an antibody described herein, or a fragment thereof.
  • a host cell contains two different vectors, a first vector comprising a polynucleotide encoding a heavy chain or a heavy chain variable region of an antibody described herein, or a fragment thereof, and a second vector comprising a polynucleotide encoding a light chain or a light chain variable region of an antibody described herein, or a fragment thereof.
  • a first host cell comprises a first vector comprising a polynucleotide encoding a heavy chain or a heavy chain variable region of an antibody described herein, or a fragment thereof
  • a second host cell comprises a second vector comprising a polynucleotide encoding a light chain or a light chain variable region of an antibody described herein.
  • a heavy chain/heavy chain variable region expressed by a first cell associated with a light chain/light chain variable region of a second cell to form an anti-CD47 antibody described herein or an antigen-binding fragment thereof.
  • a population of host cells comprising such first host cell and such second host cell.
  • a population of vectors comprising a first vector comprising a polynucleotide encoding a light chain/light chain variable region of an anti-CD47 antibody described herein, and a second vector comprising a polynucleotide encoding a heavy chain/heavy chain variable region of an anti-CD47 antibody described herein.
  • host-expression vector systems can be utilized to express antibody molecules described herein (see, e.g., U.S. Patent No. 5,807,715).
  • host-expression systems represent vehicles by which the coding sequences of interest can be produced and subsequently purified, but also represent cells which can, when transformed or transfected with the appropriate nucleotide coding sequences, express an antibody molecule described herein in situ.
  • These include but are not limited to microorganisms such as bacteria ⁇ e.g., E. coli and B.
  • subtilis transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing antibody coding sequences; yeast ⁇ e.g., Saccharomyces Pichia) transformed with recombinant yeast expression vectors containing antibody coding sequences; insect cell systems infected with recombinant virus expression vectors ⁇ e.g., baculovirus) containing antibody coding sequences; plant cell systems ⁇ e.g., green algae such as Chlamydomonas reinhardtii) infected with recombinant virus expression vectors ⁇ e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors ⁇ e.g., Ti plasmid) containing antibody coding sequences; or mammalian cell systems ⁇ e.g., COS, CHO, BHK, MDCK, HEK 293, NSO, PER.C6, VERO, CRL7030
  • cells for expressing antibodies described herein or an antigen-binding fragment thereof are CHO cells, for example CHO cells from the CHO GS SystemTM (Lonza).
  • a mammalian expression vector is pOptiVECTM or pcDNA3.3.
  • bacterial cells such as Escherichia coli, or eukaryotic cells (e.g., mammalian cells), especially for the expression of whole recombinant antibody molecule, are used for the expression of a recombinant antibody molecule.
  • mammalian cells such as Chinese hamster ovary (CHO) cells
  • CHO Chinese hamster ovary
  • a vector such as the major intermediate early gene promoter element from human cytomegalovirus
  • antibodies described herein are produced by CHO cells or NSO cells.
  • the expression of nucleotide sequences encoding antibodies described herein which immunospecifically bind to CD47 is regulated by a constitutive promoter, inducible promoter or tissue specific promoter.
  • a number of expression vectors can be advantageously selected depending upon the use intended for the antibody molecule being expressed.
  • vectors which direct the expression of high levels of fusion protein products that are readily purified can be desirable.
  • Such vectors include, but are not limited to, the E. coli expression vector pUR278 (Ruther et al., 1983, EMBO 12: 1791), in which the antibody coding sequence can be ligated individually into the vector in frame with the lac Z coding region so that a fusion protein is produced; pIN vectors (Inouye & Inouye, 1985, Nucleic Acids Res.
  • pGEX vectors can also be used to express foreign polypeptides as fusion proteins with glutathione 5-transferase (GST).
  • GST glutathione 5-transferase
  • fusion proteins are soluble and can easily be purified from lysed cells by adsorption and binding to matrix glutathione agarose beads followed by elution in the presence of free glutathione.
  • the pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety.
  • Autographa californica nuclear polyhedrosis virus (AcNPV), for example, can be used as a vector to express foreign genes.
  • the virus grows in Spodoptera frugiperda cells.
  • the antibody coding sequence can be cloned individually into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter).
  • AcNPV Autographa californica nuclear polyhedrosis virus
  • mammalian host cells a number of viral-based expression systems can be utilized.
  • the antibody coding sequence of interest can be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence.
  • This chimeric gene can then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non-essential region of the viral genome (e.g., region El or E3) will result in a recombinant virus that is viable and capable of expressing the antibody molecule in infected hosts (e.g., see Logan & Shenk, 1984, Proc. Natl. Acad. Sci. USA 8 1 :355-359).
  • Specific initiation signals can also be required for efficient translation of inserted antibody coding sequences. These signals include the ATG initiation codon and adjacent sequences. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression can be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see, e.g., Bittner et al, 1987, Methods in Enzymol. 153 :51-544).
  • a host cell strain can be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products can be important for the function of the protein.
  • Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed.
  • eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, and
  • Such mammalian host cells include but are not limited to CHO, VERO, BHK, Hela, COS, MDCK, HEK 293, NIH 3T3, W138, BT483, Hs578T, HTB2, BT20 and T47D, NS0 (a murine myeloma cell line that does not endogenously produce any immunoglobulin chains), CRL7030 and HsS78Bst cells.
  • anti- CD47 antibodies described herein e.g., an antibody comprising the CDRs of any one of antibodies Ab235-Ab255
  • mammalian cells such as CHO cells.
  • stable expression cells For long-term, high-yield production of recombinant proteins, stable expression cells can be generated.
  • cell lines which stably express an anti-CD47 antibody described herein or an antigen-binding fragment thereof can be engineered.
  • a cell provided herein stably expresses a light chain/light chain variable domain and a heavy chain/heavy chain variable domain which associate to form an antibody described herein or an antigen-binding fragment thereof.
  • host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker.
  • appropriate expression control elements e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.
  • engineered cells can be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media.
  • the selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines.
  • Such engineered cell lines can be particularly useful in screening and evaluation of compositions that interact directly or indirectly with the antibody molecule.
  • a number of selection systems can be used, including but not limited to, the herpes simplex virus thymidine kinase (Wigler et al, 1977, Cell 11 :223), hypoxanthineguanine phosphoribosyltransferase (Szybalska & Szybalski, 1992, Proc. Natl. Acad. Sci. USA 48:202), and adenine phosphoribosyltransferase (Lowy et al, 1980, Cell 22:8-17) genes can be employed in tk-, hgprt- or aprt-cells, respectively.
  • antimetabolite resistance can be used as the basis of selection for the following genes: dhfr, which confers resistance to methotrexate (Wigler et al, 1980, Natl. Acad. Sci. USA 77:357; O'Hare et al, 1981, Proc. Natl. Acad. Sci. USA
  • the host cell can be co-transfected with two or more expression vectors described herein, the first vector encoding a heavy chain derived polypeptide and the second vector encoding a light chain derived polypeptide.
  • the two vectors can contain identical selectable markers which enable equal expression of heavy and light chain polypeptides.
  • the host cells can be co-transfected with different amounts of the two or more expression vectors.
  • host cells can be transfected with any one of the following ratios of a first expression vector and a second expression vector: 1 : 1, 1 :2, 1 :3, 1 :4, 1 :5, 1 :6, 1 :7, 1 :8, 1 :9, 1 : 10, 1 : 12, 1 : 15, 1 :20, 1 :25, 1 :30, 1 :35, 1 :40, 1 :45, or 1 :50.
  • a single vector can be used which encodes, and is capable of expressing, both heavy and light chain polypeptides.
  • the light chain should be placed before the heavy chain to avoid an excess of toxic free heavy chain (Proudfoot, 1986, Nature 322:52; and Kohler, 1980, Proc. Natl. Acad. Sci. USA 77:2197-2199).
  • the coding sequences for the heavy and light chains can comprise cDNA or genomic DNA.
  • the expression vector can be monocistronic or multicistronic.
  • a multicistronic nucleic acid construct can encode 2, 3, 4, 5, 6, 7, 8, 9, 10 or more, or in the range of 2-5, 5-10 or 10-20 genes/nucleotide sequences.
  • a bicistronic nucleic acid construct can comprise in the following order a promoter, a first gene (e.g., heavy chain of an antibody described herein), and a second gene and (e.g., light chain of an antibody described herein).
  • the transcription of both genes can be driven by the promoter, whereas the translation of the mRNA from the first gene can be by a cap-dependent scanning mechanism and the translation of the mRNA from the second gene can be by a cap-independent mechanism, e.g., by an IRES.
  • immunoglobulin molecule for example, by chromatography (e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column
  • chromatography e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column
  • antibodies described herein can be fused to heterologous polypeptide sequences described herein or otherwise known in the art to facilitate purification.
  • an antibody or antigen-binding fragment described herein is isolated or purified.
  • an isolated antibody is one that is substantially free of other antibodies with different antigenic specificities than the isolated antibody.
  • a preparation of an antibody described herein is substantially free of cellular material and/or chemical precursors. The language "substantially free of cellular material” includes preparations of an antibody in which the antibody is separated from cellular components of the cells from which it is isolated or recombinantly produced.
  • an antibody that is substantially free of cellular material includes preparations of antibody having less than about 30%, 20%, 10%, 5%, 2%, 1%, 0.5%, or 0.1% (by dry weight) of heterologous protein (also referred to herein as a "contaminating protein") and/or variants of an antibody, for example, different post-translational modified forms of an antibody or other different versions of an antibody (e.g., antibody fragments).
  • heterologous protein also referred to herein as a "contaminating protein”
  • variants of an antibody for example, different post-translational modified forms of an antibody or other different versions of an antibody (e.g., antibody fragments).
  • the antibody is recombinantly produced, it is also generally substantially free of culture medium, i.e., culture medium represents less than about 20%, 10%, 2%, 1%, 0.5%, or 0.1% of the volume of the protein preparation.
  • the antibody When the antibody is produced by chemical synthesis, it is generally substantially free of chemical precursors or other chemicals, i.e., it is separated from chemical precursors or other chemicals which are involved in the synthesis of the protein. Accordingly, such preparations of the antibody have less than about 30%, 20%, 10%, or 5% (by dry weight) of chemical precursors or compounds other than the antibody of interest.
  • antibodies described herein are isolated or purified.
  • compositions, pharmaceutical compositions, and kits comprising one or more antibodies (e.g., anti-CD47 antibodies) described herein, or antigen-binding fragments thereof, or conjugates thereof.
  • compositions (e.g., pharmaceutical compositions) described herein can be for in vitro, in vivo, or ex vivo uses.
  • uses include uses to modulate ⁇ e.g., inhibit or induce/enhance) CD47 activity and uses to manage or treat a disorder, for example, cancer.
  • a pharmaceutical composition comprising an antibody ⁇ e.g., a humanized antibody) described herein (or an antigen-binding fragment thereof) and a pharmaceutically acceptable carrier or excipient.
  • the term "pharmaceutically acceptable” means being approved by a regulatory agency of the Federal or a state government, or listed in the U.S. Pharmacopeia, European Pharmacopeia or other generally recognized Pharmacopeia for use in animals, and more particularly in humans.
  • Formulations containing one or more antibodies provided herein or an antigen- binding fragment thereof can be prepared for storage by mixing the antibody having the desired degree of purity with optional physiologically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences (1990) Mack Publishing Co., Easton, PA; Remington: The Science and Practice of Pharmacy, 21st ed. (2006) Lippincott Williams & Wilkins,
  • Such formulations can, for example, be in the form of, e.g., lyophilized formulations or aqueous solutions.
  • Pharmaceutical carriers suitable for administration of the antibodies provided herein include any such carriers known to those skilled in the art to be suitable for the particular mode of administration. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; and/or non-ionic surfactants such as TWEENTM, PLURONICSTM or polyethylene glycol (PEG).
  • Formulations to be used for in vivo administration can be sterile. This can be readily accomplished, for example, by filtration through, e.g., sterile filtration membranes.
  • compositions provided herein contain therapeutically effective amounts of one or more of the antibodies or antigen-binding fragments provided herein in a pharmaceutically acceptable carrier.
  • Such pharmaceutical compositions are useful in the prevention, treatment, management or amelioration of a condition or disorder described herein or one or more symptoms thereof.
  • compositions provided herein can contain one or more antibodies provided herein or an antigen-binding fragment thereof.
  • compositions are provided wherein antibodies or antigen-binding fragments described herein are formulated into suitable pharmaceutical preparations, such as solutions, suspensions, powders, sustained release formulations or elixirs in sterile solutions or suspensions for parenteral administration, or as transdermal patch preparation and dry powder inhalers.
  • compositions provided herein are formulated for single dosage administration.
  • the weight fraction of compound is dissolved, suspended, dispersed or otherwise mixed in a selected carrier at an effective concentration such that the treated condition is relieved, prevented, or one or more symptoms are ameliorated.
  • an antibody provided herein is included in the pharmaceutically acceptable carrier in an effective amount sufficient to exert a therapeutically useful effect in the absence of, or with minimal or negligible, undesirable side effects on the patient treated.
  • compositions provided herein will depend on, e.g., the physicochemical characteristics of the antibody, the dosage schedule, and amount administered as well as other factors.
  • compositions described herein are provided for administration to humans or animals (e.g., mammals) in unit dosage forms, such as sterile parenteral (e.g., intravenous) solutions or suspensions containing suitable quantities of the compounds or pharmaceutically acceptable derivatives thereof.
  • Pharmaceutical compositions are also provided for administration to humans and animals in unit dosage form, such as tablets, capsules, pills, powders, granules, and oral or nasal solutions or suspensions, and oil-water emulsions containing suitable quantities of an anti-CD47 antibody or pharmaceutically acceptable derivatives thereof.
  • the antibody is, in one embodiment, formulated and administered in unit-dosage forms or multiple-dosage forms.
  • Unit-dose forms as used herein refers to physically discrete units suitable for human or animal (e.g., mammal) subjects and packaged individually. Each unit-dose contains a predetermined quantity of an anti-CD47 antibody sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carrier, vehicle or diluent. Examples of unit-dose forms include ampoules and syringes and individually packaged tablets or capsules. Unit-dose forms can be administered in fractions or multiples thereof.
  • a multiple- dose form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dose form. Examples of multiple-dose forms include vials, bottles of tablets or capsules or bottles. Hence, in specific aspects, multiple dose form is a multiple of unit-doses which are not segregated in packaging.
  • one or more anti-CD47 antibodies described herein or an antigen-binding fragment thereof are in a liquid pharmaceutical formulation. Liquid
  • compositions can, for example, be prepared by dissolving, dispersing, or otherwise mixing an antibody and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, glycols, and the like, to thereby form a solution or suspension.
  • a pharmaceutical composition provided herein to be administered can also contain minor amounts of nontoxic auxiliary substances such as wetting agents, emulsifying agents, solubilizing agents, and pH buffering agents and the like.
  • Parenteral administration in one embodiment, is characterized by injection, either subcutaneously, intramuscularly or intravenously is also contemplated herein.
  • injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions.
  • the injectables, solutions and emulsions also contain one or more excipients. Suitable excipients are, for example, water, saline, dextrose, glycerol or ethanol.
  • Other routes of administration may include, enteric administration, intracerebral administration, nasal administration, intraarterial administration, intracardiac administration, intraosseous infusion, intrathecal administration, and intraperitoneal administration.
  • Preparations for parenteral administration include sterile solutions ready for injection, sterile dry soluble products, such as lyophilized powders, ready to be combined with a solvent just prior to use, including hypodermic tablets, sterile suspensions ready for injection, sterile dry insoluble products ready to be combined with a vehicle just prior to use and sterile emulsions.
  • the solutions can be either aqueous or nonaqueous.
  • suitable carriers include physiological saline or phosphate buffered saline (PBS), and solutions containing thickening and solubilizing agents, such as glucose, polyethylene glycol, and polypropylene glycol and mixtures thereof.
  • suitable carriers include physiological saline or phosphate buffered saline (PBS), and solutions containing thickening and solubilizing agents, such as glucose, polyethylene glycol, and polypropylene glycol and mixtures thereof.
  • Pharmaceutically acceptable carriers used in parenteral preparations include aqueous vehicles, nonaqueous vehicles, antimicrobial agents, isotonic agents, buffers, antioxidants, local anesthetics, suspending and dispersing agents, emulsifying agents, sequestering or chelating agents and other pharmaceutically acceptable substances.
  • Pharmaceutical carriers also include ethyl alcohol, polyethylene glycol and propylene glycol for water miscible vehicles; and sodium hydroxide, hydrochloric acid, citric acid or lactic acid for pH adjustment.
  • intravenous or intraarterial infusion of a sterile aqueous solution containing an anti-CD47 antibody or fragment described herein is an effective mode of administration.
  • Another embodiment is a sterile aqueous or oily solution or suspension containing an anti-CD47 antibody described herein injected as necessary to produce the desired pharmacological effect.
  • an anti-CD47 antibody described herein can be suspended in micronized or other suitable form.
  • the form of the resulting mixture depends upon a number of factors, including the intended mode of administration and the solubility of the compound in the selected carrier or vehicle.
  • the pharmaceutical formulations are lyophilized powders, which can be reconstituted for administration as solutions, emulsions and other mixtures. They can also be reconstituted and formulated as solids or gels.
  • Lyophilized powder can, for example, be prepared by dissolving an anti-CD47 antibody provided herein, in a suitable solvent.
  • the lyophilized powder is sterile.
  • Suitable solvents can contain an excipient which improves the stability or other pharmacological component of the powder or reconstituted solution, prepared from the powder. Excipients that can be used include, but are not limited to, dextrose, sorbital, fructose, corn syrup, xylitol, glycerin, glucose, sucrose or other suitable agent.
  • a suitable solvent can also contain a buffer, such as citrate, sodium or potassium phosphate or other such buffer known to those of skill in the art at, in one embodiment, about neutral pH.
  • sterile filtration of the solution followed by lyophilization under standard conditions known to those of skill in the art provides an example of a formulation.
  • the resulting solution will be apportioned into vials for lyophilization.
  • Lyophilized powder can be stored under appropriate conditions, such as at about 4 °C to room temperature.
  • Reconstitution of this lyophilized powder with water for injection provides a formulation for use in parenteral administration.
  • the lyophilized powder is added to sterile water or other suitable carrier.
  • anti-CD47 antibodies provided herein can be formulated for local administration or topical application, such as for topical application to the skin and mucous membranes, such as in the eye, in the form of gels, creams, and lotions and for application to the eye or for intraci sternal or intraspinal application.
  • Topical administration is contemplated for transdermal delivery and also for administration to the eyes or mucosa, or for inhalation therapies.
  • Nasal solutions of the active compound alone or in combination with other pharmaceutically acceptable excipients can also be administered.
  • Anti-CD47 antibodies and other compositions provided herein can also be formulated to be targeted to a particular tissue, organ, or other area of the body of the subject to be treated. Many such targeting methods are well known to those of skill in the art. All such targeting methods are contemplated herein for use in the instant compositions. For non-limiting examples of targeting methods, see, e.g., U.S. Patent Nos.
  • anti-CD47 antibodies described herein are targeted (or otherwise administered) to the visual organs, bone marrow, gastrointestinal tract, lungs, brain, or joints.
  • an anti-CD47 antibody described herein is capable of crossing the blood-brain barrier.
  • a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions described herein, such as one or more anti-CD47 antibodies provided herein.
  • Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • kits comprising one or more of the antibodies or antibody fragments described herein.
  • a kit comprises an antibody or antibody fragment described herein, in one or more containers.
  • kits described herein contain a substantially purified CD47 antigen as a control.
  • the kits described herein further comprise a control antibody which does not react with a CD47 antigen.
  • kits described herein contain one or more elements for detecting the binding of a modified antibody to a CD47 antigen (e.g., the antibody can be conjugated to a detectable substrate such as a fluorescent compound, an enzymatic substrate, a radioactive compound or a luminescent compound, or a second antibody which recognizes the first antibody can be conjugated to a detectable substrate).
  • a detectable substrate such as a fluorescent compound, an enzymatic substrate, a radioactive compound or a luminescent compound, or a second antibody which recognizes the first antibody can be conjugated to a detectable substrate.
  • a kit provided herein can include a recombinantly produced or chemically synthesized CD47 antigen.
  • the CD47 antigen provided in the kit can also be attached to a solid support.
  • the detecting means of the above described kit includes a solid support to which a CD47 antigen is attached.
  • Such a kit can also include a non-attached reporter-labeled anti-human antibody or anti-mouse/rat antibody. In this embodiment, binding of the antibody to the CD47 antigen can be detected by binding of the said reporter-labeled antibody.
  • provided herein are methods of inhibiting (e.g., partially inhibiting) a CD47 activity with an anti-CD47 antibody described herein.
  • provided herein are methods of managing or treating a condition or disorder, such as cancer, using an anti-CD47 antibody described herein. In certain embodiments, provided herein are methods of protecting against a condition or disorder, such as cancer, using an anti- CD47 antibody described herein.
  • provided herein are methods for managing, treating, preventing or protecting against cancer, comprising administering to a subject in need thereof a therapeutically effective amount of an antibody or an antigen-binding fragment described herein that binds specifically to CD47 (e.g., human CD47).
  • a method of alleviating, inhibiting or reducing the progression or severity of one or more symptoms associated with cancer comprising administering to a subject in need thereof a therapeutically effective amount of an antibody or an antigen-binding fragment described herein that binds specifically to CD47 (e.g., human CD47).
  • administer refers to the act of injecting or otherwise physically delivering a substance (e.g., a humanized anti-CD47 antibody provided herein or an antigen-binding fragment thereof) to a subject or a patient (e.g., human), such as by mucosal, topical, intradermal, parenteral, intravenous, subcutaneous, intramuscular delivery and/or any other method of physical delivery described herein or known in the art.
  • a substance e.g., a humanized anti-CD47 antibody provided herein or an antigen-binding fragment thereof
  • the terms "effective amount” or “therapeutically effective amount” refer to an amount of a therapy (e.g., an antibody or pharmaceutical composition provided herein) which is sufficient to reduce and/or ameliorate the severity and/or duration of a given condition, disorder or disease (e.g., cancer, metastasis, or angiogenesis) and/or a symptom related thereto. These terms also encompass an amount necessary for the reduction, slowing, or amelioration of the advancement or progression of a given disease, reduction, slowing, or amelioration of the recurrence, development or onset of a given disease, and/or to improve or enhance the
  • an effective amount as used herein also refers to the amount of an antibody described herein to achieve a specified result.
  • the term "in combination” in the context of the administration of other therapies refers to the use of more than one therapy.
  • the use of the term “in combination” does not restrict the order in which therapies are administered.
  • the therapies may be
  • the terms “manage,” “managing,” and “management” refer to the beneficial effects that a subject derives from a therapy (e.g., a prophylactic or therapeutic agent), which does not result in a cure of a condition associated with CD47.
  • a subject is administered one or more therapies (e.g., prophylactic or therapeutic agents, such as an antibody described herein) to "manage” a condition or disorder described herein, one or more symptoms thereof, so as to prevent the progression or worsening of the condition or disorder.
  • therapies e.g., prophylactic or therapeutic agents, such as an antibody described herein
  • the terms "impede” or “impeding” in the context of a condition or disorder provided herein refer to the total or partial inhibition (e.g., less than 100%, 95%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, or 5%) or blockage of the development, recurrence, onset or spread of a condition or disorder provided herein (e.g., cancer, metastasis, or angiogenesis) and/or symptom related thereto, resulting from the administration of a therapy or combination of therapies provided herein (e.g., a combination of prophylactic or therapeutic agents, such as an antibody described herein).
  • a therapy or combination of therapies provided herein (e.g., a combination of prophylactic or therapeutic agents, such as an antibody described herein).
  • a subject is a mammal such as a non-primate (e.g., cows, pigs, horses, cats, dogs, goats, rabbits, rats, mice, etc.) or a primate (e.g., monkey and human), for example a human.
  • the subject is a mammal, e.g., a human, diagnosed with a condition or disorder provided herein (e.g., cancer, metastasis, or angiogenesis).
  • the subject is a mammal, e.g., a human, at risk of developing a condition or disorder provided herein (e.g., cancer, metastasis, or angiogenesis).
  • the subject is human.
  • CD47 is amplified in cells of a subject, e.g., the human subject.
  • Identification of cd47 amplification in a sample from a subject can be performed by assays known to one of ordinary skill in the art, such as, e.g., quantitative reverse transcription PCR, immunoblot assays, DNA fingerprinting, karyotyping (for example, by multicolor fluorescence in situ hybridization (mFISH)), comparative genome hybridization, and gene expression profiling.
  • protein expression of tumor samples can be characterized using immunohistochemical assays to measure the amount of CD47 protein present in a sample.
  • Identification of mutations or deltions in a sample from a subject can be performed by assays known to one of ordinary skill in the art, such as, e.g., DNA extraction, generation of complementary DNA, and cDNA sequencing.
  • the cDNA sequence for example, can be utilized to obtain the translation product by methods known to one of ordinary skill in the art.
  • Genetic deletions and amino acid substitutions can be identified by, for example, comparing the sequence from the sample from the subject to a a wild type and/or consensus sequence.
  • CD47 is amplified in the subject treated in accordance with the methods provided herein.
  • Identification of CD47 amplification in a sample from a subject is performed by assays known to one of ordinary skill in the art, such as, e.g., quantitative reverse transcription PCR or immunoblot assays.
  • Identification of mutations or deletions in a sample from a subject are performed by assays known to one of ordinary skill in the art, such as, e.g., DNA extraction, generation of complementary DNA, and cDNA sequencing.
  • the cDNA sequence for example, is utilized to obtain the translation product by methods known to one of ordinary skill in the art.
  • the terms “therapies” and “therapy” can refer to any protocol(s), method(s), compositions, formulations, and/or agent(s) that can be used in the prevention, treatment, management, or amelioration of a condition or disorder or symptom thereof (e.g., a condition or disorder provided herein (e.g., cancer) or one or more symptoms or condition associated therewith).
  • a condition or disorder or symptom thereof e.g., a condition or disorder provided herein (e.g., cancer) or one or more symptoms or condition associated therewith.
  • the terms “therapies” and “therapy” refer to drug therapy, adjuvant therapy, radiation, surgery, biological therapy, supportive therapy, and/or other therapies useful in treatment, management, prevention, or amelioration of a condition or disorder or one or more symptoms thereof (e.g., cancer or one or more symptoms or condition associated therewith).
  • the term “therapy” refers to a therapy other than an anti- CD47 antibody described herein or pharmaceutical composition thereof.
  • an "additional therapy” and “additional therapies” refer to a therapy other than a treatment using an anti-CD47 antibody described herein or pharmaceutical composition thereof.
  • a therapy includes the use of an anti-CD47 antibody described herein as an adjuvant therapy. For example, using an anti-CD47 antibody described herein in conjunction with a drug therapy, biological therapy, surgery, and/or supportive therapy.
  • hematological cancer refers to a cancer of the blood, and includes leukemia, lymphoma and myeloma among others.
  • Leukemia refers to a cancer of the blood in which too many white blood cells that are ineffective in fighting infection are made, thus crowding out the other parts that make up the blood, such as platelets and red blood cells. It is understood that cases of leukemia are classified as acute or chronic.
  • leukemia include, by way of non-limiting example, acute lymphocytic leukemia (ALL); acute myeloid leukemia (AML); chronic lymphocytic leukemia (CLL); chronic myelogenous leukemia (CML); Myeloproliferative disorder/neoplasm (MPDS); and myelodysplasia syndrome.
  • ALL acute lymphocytic leukemia
  • AML acute myeloid leukemia
  • CLL chronic lymphocytic leukemia
  • CML chronic myelogenous leukemia
  • MPDS Myeloproliferative disorder/neoplasm
  • myelodysplasia syndrome myelodysplasia syndrome.
  • Lymphoma may refer to a Hodgkin's lymphoma, both indolent and aggressive non-Hodgkin' s lymphoma, Burkitt' s lymphoma, and follicular lymphoma (small cell and large cell), among others.
  • Myeloma may refer to multiple myeloma (MM), giant cell myeloma, heavy-chain myeloma, and light chain or Bence-Jones myeloma.
  • MM multiple myeloma
  • giant cell myeloma giant cell myeloma
  • heavy-chain myeloma heavy-chain myeloma
  • light chain or Bence-Jones myeloma may refer to multiple myeloma (MM), giant cell myeloma, heavy-chain myeloma, and light chain or Bence-Jones myeloma.
  • Non-limiting examples of a condition which can be treated or managed with an anti- CD47 antibody described herein include hematological caner and/or solid tumors.
  • Hematological cancers include, e.g., leukemia, lymphoma and myeloma.
  • leukemia include, by way of non-limiting example, acute lymphocytic leukemia (ALL); acute myeloid leukemia (AML); chronic lymphocytic leukemia (CLL); chronic myelogenous leukemia (CML); Myeloproliferative disorder/neoplasm (MPDS); and myelodysplasia syndrome.
  • ALL acute lymphocytic leukemia
  • AML acute myeloid leukemia
  • CLL chronic lymphocytic leukemia
  • CML chronic myelogenous leukemia
  • MPDS Myeloproliferative disorder/neoplasm
  • myelodysplasia syndrome myelodysplasia syndrome.
  • lymphoma include, by way of non-limiting example, Hodgkin's lymphoma, both indolent and aggressive non-Hodgkin's lymphoma, Burkitt's lymphoma, and follicular lymphoma (small cell and large cell).
  • Certain forms of myeloma include, by way of non-limiting example, multiple myeloma (MM), giant cell myeloma, heavy-chain myeloma, and light chain or Bence- Jones myeloma.
  • Solid tumors include, e.g., breast tumors, ovarian tumors, lung tumors, pancreatic tumors, prostate tumors, melanoma tumors, colorectal tumors, lung tumors, head and neck tumors, bladder tumors, esophageal tumors, liver tumors, and kidney tumors.
  • Symptoms associated with cancers and other neoplastic disorders include, for example, inflammation, fever, general malaise, fever, pain, often localized to the inflamed area, loss of appetite, weight loss, edema, headache, fatigue, rash, anemia, muscle weakness, muscle fatigue and abdominal symptoms such as, for example, abdominal pain, diarrhea or constipation.
  • anti-CD47 antibodies useful in treating, delaying the progression of, impeding, preventing relapse of or alleviating a symptom of a cancer (e.g., MM, NHL, AML, breast cancer, bladder cancer, non-small cell lung cancer/carcinoma, hepatocellular carcinoma (HCC), sarcoma, and head and neck cancer).
  • a cancer e.g., MM, NHL, AML, breast cancer, bladder cancer, non-small cell lung cancer/carcinoma, hepatocellular carcinoma (HCC), sarcoma, and head and neck cancer.
  • the CD47 antibodies described herein are useful in treating hematological malignancies and/or tumors, e.g., hematological malignancies and/or tumors.
  • the CD47 antibodies described herein are useful in treating CD47+ tumors.
  • the CD47 antibodies described herein are useful in treating non-Hodgkin's lymphoma (NHL), acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), multiple myeloma (MM), breast cancer, ovarian cancer, head and neck cancer, bladder cancer, melanoma, colorectal cancer, pancreatic cancer, lung cancer, leiomyoma, leiomyosarcoma, glioma, glioblastoma, and so on.
  • NHL non-Hodgkin's lymphoma
  • ALL acute lymphocytic leukemia
  • AML acute myeloid leukemia
  • CLL chronic lymphocytic leukemia
  • CML chronic myelogenous leukemia
  • MM multiple myeloma
  • breast cancer ovarian cancer
  • head and neck cancer bladder cancer
  • melanoma
  • Solid tumors include, e.g., breast tumors, ovarian tumors, lung tumors (e.g., NSCLC), pancreatic tumors, prostate tumors, melanoma tumors, colorectal tumors, lung tumors, head and neck tumors, bladder tumors, esophageal tumors, liver tumors (e.g., hepatocellular carcinoma), sarcoma, and kidney tumors.
  • lung tumors e.g., NSCLC
  • pancreatic tumors e.g., prostate tumors, melanoma tumors, colorectal tumors, lung tumors, head and neck tumors, bladder tumors, esophageal tumors, liver tumors (e.g., hepatocellular carcinoma), sarcoma, and kidney tumors.
  • liver tumors e.g., hepatocellular carcinoma
  • sarcoma e.g., hepatocellular carcinoma
  • a method of treating cancer e.g., a hematological disorder/cancer or solid cancer
  • administering e.g., administering concurrently or sequentially
  • another anti-cancer agent e.g., another anti-cancer agent.
  • the anti-cancer agent is a chermotherapeutic agent (e.g., microtubule disassembly blocker, antimetabolite, topisomerase inhibitor, and DNA crosslinker or damaging agent).
  • the anti-cancer agent is a tyrosine kinase inhibitor (e.g., GLEEVEC® (imatinib mesylate) or
  • SUTENT® (SU1 1248 or Sunitinib)
  • Other non-limiting examples of tyrosine kinse inhibitors include 706 and AMNI07 (nilotinib).
  • a method of treating cancer e.g., a hematological disorder/cancer or solid cancer
  • administering e.g., administering concurrently or sequentially
  • an anti-CD47 antibody described herein or antigen-binding fragment thereof which specifically binds to CD47 such as human CD47
  • radiation therapy e.g., radiation therapy
  • a method of promoting e.g., inducing or increasing phagocytosis, e.g., macrophage mediated phagocytic killing of tumor cells, comprising contacting an effective amount of an anti-CD47 antibody described herein which specifically binds to human CD47 with tumor cells.
  • Also provided herein is a method of promoting (e.g., inducing or increasing) phagocytosis, e.g., macrophage mediated phagocytic killing of tumor cells, in a subject in need thereof (e.g., a subject with tumor cells, such as tumor cells expressing CD47), comprising administering to the subject an effective amount of an anti- CD47 antibody described herein which specifically binds to human CD47.
  • a subject in need thereof e.g., a subject with tumor cells, such as tumor cells expressing CD47
  • a method of reducing tumor volumn comprising contacting an effective amount of an anti-CD47 antibody described herein which specifically binds to human CD47 with the tumor. Also provided herein is a method of reducing tumor volumn in a subject in need thereof (e.g., a subject with a tumor, such as a CD47 expressing tumor), comprising administering to the subject an effective amount of an anti-CD47 antibody described herein which specifically binds to human CD47.
  • a method of inhibiting cancer cell growth or proliferation comprising contacting an effective amount of an anti-CD47 antibody described herein which specifically binds to human CD47 with cancer cells. Also provided herein is a method of inhibiting cancer cell growth or proliferation in a subject in need thereof (e.g., a subject with cancer cells, such as CD47 expressing cancer cells), comprising administering to the subject an effective amount of an anti-CD47 antibody described herein which specifically binds to human CD47.
  • anti-CD47 antibodies described herein and antigen-binding fragments thereof, which specifically bind to an ECD of human CD47 can be used for diagnostic purposes to detect, diagnose, or monitor a condition described herein (e.g., a condition involving CD47 and/or abnormal CD47 signaling and/or abnormal CD47 expression), such as cancer (e.g., colorectal cancer, gastric cancer, lung cancer, or melanoma).
  • a condition described herein e.g., a condition involving CD47 and/or abnormal CD47 signaling and/or abnormal CD47 expression
  • cancer e.g., colorectal cancer, gastric cancer, lung cancer, or melanoma
  • anti- CD47 antibodies described herein or an antigen-binding fragment thereof for use in diagnostic purposes are labeled.
  • Methods provided herein for diagnostic purposes to detect, diagnose, or monitor a condition described herein can be in vitro methods, in situ methods, or ex vivo methods. Methods provided herein for diagnostic purposes to detect, diagnose,
  • kits for the detection of a condition described herein, such as cancer comprising: (a) assaying the expression of CD47 in a sample of a subject using one or more antibodies described herein or an antigen-binding fragment thereof; and (b) comparing the level of CD47 expression with a control level, e.g., levels in normal tissue samples (e.g., from a patient not having a condition described herein, or from the same patient before onset of the condition), whereby an increase or decrease in the assayed level of CD47 expression compared to the control level of CD47 expression is indicative of a condition described herein.
  • a control level e.g., levels in normal tissue samples (e.g., from a patient not having a condition described herein, or from the same patient before onset of the condition)
  • kits for the detection of cancer expressing CD47 comprising: (a) assaying the expression of CD47 in a sample of a subject using one or more antibodies described herein or an antigen-binding fragment thereof; and (b) comparing the level of CD47 expression with a control level, e.g., levels in normal samples (e.g., from a patient not having cancer, a patient having cancer that does not overexpress CD47, or from the same patient before onset of cancer).
  • a control level e.g., levels in normal samples (e.g., from a patient not having cancer, a patient having cancer that does not overexpress CD47, or from the same patient before onset of cancer).
  • a control level e.g., levels in normal samples (e.g., from a patient not having cancer, a patient having cancer that does not overexpress CD47, or from the same patient before onset of cancer).
  • a control level e.g., levels in normal samples (e.g., from a patient not
  • a method of diagnosing a CD47- expressing cancer in a patient comprising the steps of:
  • a method of monitoring CD47 protein level during treatment of a CD47-expressing cancer in a patient comprising the steps of:
  • sample e.g., bodily fluid or tissue sample
  • samples which can be used in diagnostic methods provided herein include, serum sample, plasma sample, tissue sample, urine sample, tumor sample, and stool sample.
  • Antibodies described herein can be used to assay CD47 levels in a biological sample using classical immunohistological methods as described herein or as known to those of skill in the art (e.g., see Jalkanen et al, 1985, J. Cell. Biol. 101 :976-985; and Jalkanen et al, 1987, J. Cell . Biol. 105:3087-3096).
  • Other antibody-based methods useful for detecting protein gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA).
  • Suitable antibody assay labels include enzyme labels, such as, glucose oxidase; radioisotopes, such as iodine (1251, 1211), carbon (14C), sulfur (35S), tritium (3H), indium (121In), and technetium (99Tc); luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine, and biotin.
  • enzyme labels such as, glucose oxidase
  • radioisotopes such as iodine (1251, 1211), carbon (14C), sulfur (35S), tritium (3H), indium (121In), and technetium (99Tc)
  • luminescent labels such as luminol
  • fluorescent labels such as fluorescein and rhodamine, and biotin.
  • monitoring of a condition described herein is carried out by repeating the method for diagnosing for a period of time after initial diagnosis.
  • Presence of the labeled molecule can be detected in the subject using methods known in the art for in vivo scanning. Skilled artisans will be able to determine the appropriate method for detecting a particular label. Methods and devices that may be used in the diagnostic methods of the invention include, but are not limited to, computed tomography (CT), whole body scan such as position emission tomography (PET), magnetic resonance imaging (MRI), and
  • Exemplary nucleotide sequences encoding the above heavy chain sequences are provided at SEQ ID Nos. 23, 26, 27, 24, 28, 29, 25, 32 and 31, respectively.
  • IgGl variant antibodies such as IgGl -5m and IgGl-13mZ, and purification were carried out.
  • the scale-up conditions are described in more details in the Tables (e.g., Tables 3-6) below. Both variants were made with essentially the same method; the most significant difference was in reaction temperature.
  • IgGl -5m was expressed at 25°C and IgGl-13mZ was run at 30°C.
  • IgGl-13mZ showed similar results at 25°C and 30°C, but IgGl- 5m showed higher titer at 25°C versus 30°C.
  • IgGl-13mZ (IgGl, VH1-18 framework, 13+2 mutations) Scale-up Expression:
  • S30 cell-free extract was treated with 50 uM iodoacetamide (IAM) for 30 minutes at room temperature. After this treatment, the extract was combined with the reagents in Table 3 and transferred to a bioreactor (Dci-BioLafitte Evo Bioreactor, 10L maximum working volume). The bioreactor controls were configured as listed in Table 4. After 6 hours of reaction, an additional 5 mM (final concentration) oxidized glutathione was added to the reaction. The oxidized glutathione was prepared as a 250 mM stock solution with the pH adjusted to between 7 and 8 before addition to the reactor. The reaction was run for a total of 15 hours before transferring to downstream processing.
  • IAM iodoacetamide
  • Anti-CD47 antibody IgGl -5m (IgGl, VH1-18 framework, 5 mutations) Scale-up Expression:
  • S30 cell-free extract was treated with 50 uM iodoacetamide (IAM) for 30 minutes at room temperature. After this treatment, the extract was combined with the reagents in Table 5 and transferred to a bioreactor (Sartorius Biostat Q Bioreactor, 500 mL maximum working volume). The bioreactor controls were configured as listed in Table 6. After 5.5 hours of reaction, an additional 5 mM (final concentration) oxidized glutathione was added to the reaction. The oxidized glutathione was prepared as a 250 mM stock solution with the pH adjusted to between 7 and 8 before addition to the reactor. The reaction was run for a total of 15.7 hours before transferring to downstream processing.
  • IAM iodoacetamide
  • Immobilization Anti-human Fc (AHC) surfaces were prepared by amine-coupling monoclonal mouse anti-human Fc IgG (included in the Biacore Human Antibody Capture Kit, GE Life Sciences Cat # BR- 1008-39) to a Biacore CM5 sensor chip surface.
  • the running buffer for the immobilization procedure for the immobilization procedure was HBS-EP+ (10 mM HEPES, 150 mM NaCl, 3 mM EDTA, 0.05% v/v P-20 as surfactant). The following was performed in all four flow cells to prepare surfaces amine-coupled anti-human Fc IgG.
  • the CM5 chip surface was activated by injecting a 1 : 1 (v/v) mixture of 400 mM EDC and 100 mM NHS for 7 minutes at 10 ⁇ ,/ ⁇ . Following this treatment, anti-human IgG was diluted to 25 ⁇ g/mL in 10 mM sodium acetate buffer pH 5.0, and injected over all the flow cells at 10 ⁇ 7 ⁇ for 7 minutes. Then, Ethanolamine was injected at 10 ⁇ ⁇ for 7 minutes to block all the surfaces. This procedure resulted in immobilization levels of -10,000 - 11,000 RU on the sensor chip.
  • CD47 analyte was injected over the flow cells for 180 seconds (3 minutes) at 50 ⁇ 7 ⁇ , and complex dissociation was monitored for 900 seconds (15 minutes). Buffer blanks were also run, and were used to reference the analyte binding data before fitting. Anti-human Fc surfaces were regenerated with two 30-second injections of 3M MgCl 2 at 30 uL/min between each analyte binding cycle.
  • Figures 2A-2E depict individual sensorgrams for antibody variants IgGl -5m (Figure 2A), IgGl-13m ( Figure 2B), IgGl-13mZ ( Figure 2C), IgG4P-5m (Figure 2D), and IgG4PE-5m ( Figure 2E); and Figure 2F depicts a sensorgram for anti-CD47 control antibody.
  • sample variants were titrated 1 :2 in FACS buffer (PBS 2% FCS) starting from a high concentration of -66 nM for each sample.
  • FACS buffer PBS 2% FCS
  • 0.1 x 10 6 CCRF-EM cells were plated in a 96 well plate and incubated in 50 ⁇ FACS buffer containing the indicated concentration of sample variants for 1 hour in ice. Cells were then washed with 150 ⁇ FACS buffer and incubated in 50 ⁇ FACS buffer containing the secondary antibody (anti-human-IgG- PE, Jackson ImmunoResearch) diluted 1 : 100, for lhr in ice.
  • secondary antibody anti-human-IgG- PE, Jackson ImmunoResearch
  • PBMCs Human peripheral blood mononuclear cells
  • buffy coats white layer between red blood cells and plasma in a unit of whole blood after it has been spun down in a centrifuge
  • Buffy coats were diluted with PBS 2-fold and layered over 15 ml NycoPrep 1.077 (Axis-Shield, Dundee, Scotland) in 50 ml Leucosep tubes (Greiner Bio One, Monroe, NC, USA) and centrifuged at 1,000 x g for 20 minutes.
  • PBMCs were collected from the interface, washed with 35 ml PBS and centrifuged at 250 x g for 5 minutes.
  • Contaminating red blood cells were lysed with 10 ml ACK Lysing Buffer (Lonza, Allendale, NJ, USA) for 2 min and cells were diluted with 40 ml PBS and passed through a 40 um cell strainer (BD Biosciences, San Jose, CA, USA). Cells were centrifuged at 250 x g for 5 mins and resuspended in 30 ml RPMI media containing 10% FBS, 2 mM glutamine and penicillin- streptomycin. PBMCs were counted and cultured at 5xl0 6 cells/ml in RPMI media overnight.
  • CD14-positive monocytes were isolated with CD14 microbeads (Miltenvi Biotech, Auburn, CA, USA) using the AutoMacs Pro and cultured in RPMI media containing 50 ng/ml M-CSF (Peprotech, Rocky Hill, NJ, USA) for 5-7 days to obtain differentiated macrophages. Ceils were frozen down in Recovery Ceil Culture Freezing Medium (Life Technologies, Grand Island, NY, USA).
  • CCRF-CEM ceils (80,000 cells in 50 ⁇ ) and anti-CD47 antibodies in 50 ⁇ PM I media were added to macrophages. Anti-CD47 antibodies were tested at a final concentration of 4 ng/ml to 10 ⁇ g/mi. Plates were briefly centrifuged and incubated at 37°C for 3 hours. Macrophages were washed 3x with PBS to remove CCRF-CEM cells and detached with 50 ⁇ Accutase (BD Biosciences, San Jose, CA, USA) at 37°C for 10 minutes. Macrophages were collected, washed once with FACS wash buffer (PBS containing 0.2% FBS) and stained with anti-CD14-APC for 15 minute.
  • FACS wash buffer PBS containing 0.2% FBS
  • hemagglutination of human red blood cells RBCs. Therefore, hemagglutination assays were carried out to characterize anti-CD47 antibodies ability to promote agglutination of RBCs.
  • Human RBCs were sourced from Alternative Research (Cat# IPLA-WB3). Human RBCs (2 mLs) were washed in 10 mLs of lx dPBS (pH 7.4) and centrifuged for 10 minutes at 500g (1500rpm). The supernatant was aspirated, and human RBCs were washed twice, resuspended in 8 mL 1 x dPBS for a 20% solution of RBCs. Dilution for the Anti-Human RBC (Rabbit) antibody (Rockland Immunochemicals Inc., Catalog #109-4139, Lot 27233), positive control, was 1 :64 with 1 :3 serial dilutions (lOx).
  • Rabbit Anti-Human RBC
  • the starting concentration for the samples was 1000 nM with 1 :3 serial dilutions (lOx).
  • Each antibody titration were pipette (50 ⁇ ) to all wells of a U-bottom 96well plate.
  • RBC solution 50 ⁇ of 20% RBC solution
  • MCA911 Mouse Anti -Human CD47 (clone BRIC 126, Abnova) served as positive controls.
  • Assays were visualized from the top of the plate. Negative (non- hemagglutination) results appear as intact red dots, while positive (hemagglutination) results appear as a dispersed red mat.
  • Results Only positive controls (rabbit anti-human RBC antibody and MCA911 (mouse anti-human CD47 antibody)) showed hemagglutination. No CHO and cell free (CF)- expressed anti-CD47 monoclonal antibodies, including IgGl -parental, IgG4P -parental, IgG4PE- parental, IgGl-13mZ-CF, IgGl-13m-CF, IgGl-5m-CF, IgG4P-5m-CF, IgG4PE-5m-CF, show evidence of hemagglutination.
  • CF cell free
  • IgGl-QNl-CHO shows activity in the CIQ ELISA assay , while IgG4P, IgG4PE, and scFv anti-CD47 monoclonal antibodies do not show activity ("NA") in CIQ ELISA assay.
  • EC 50 values are presented in the table below (Table 10).
  • CD47-expressing cell lines were harvested and re- suspended in CDC buffer (RPMI 1640, L-glutamine (lOOx stock), and 1% BSA) at 0.3 million cells per mL. Cells were then plated at 10,000 cells per well in a 96 well white tissue culture plate (Falcon) and incubated with sample anti-CD47 antibody variants at a final concentration of 10 ⁇ g/mL in CDC buffer at 37°C for 1 hour. Spin filters (Costar SpinX microcentrifuge tubes) were used to remove residual contaminants. Rabbit (7.5%) or human (30%) serum were then added at a final concentration of 2.5% and 10%, respectively and incubate for 2 hours at 37°C.
  • CDC buffer RPMI 1640, L-glutamine (lOOx stock), and 1% BSA
  • PBC were prepared from buffy coats. Buffy coats were diluted with PBS 2- fold and layered over 15 ml NycoPrep 1.077 (Axis-Shield, Dundee, Scotland) in 50 ml Leucosep tubes (Greiner Bio One, Monroe, NC, USA) and centrifuged at 1,000 x g for 20 minutes. PBMCs were collected from the interface and washed with 35 ml PBS and centrifuged at 250 x g for 5 minutes.
  • Contaminating red blood cells were lysed with 10 ml ACK Lysing Buffer (Lonza, Allendale, NJ, USA) for 2 minutes and cells were diluted with 40 ml PBS and passed through a 40 ⁇ cell strainer (BD Biosciences, San Jose, CA, USA). Cells were centrifuged at 250 x g for 5 minutes and resuspended in 30 ml RPMI media containing 10% FBS, 2 mM glutamine and penicillin-streptomycin. 10,000 CCRF-CEM or SKBR3 cells were co-cultured with 300,000 PBMCs per each well in a 96 well U bottom polypropylene plate.
  • PBMC peripheral blood center
  • Three-fold dilutions of the sample variants were added to each well in duplicates, starting from a highest concentration of 22.2 nM and incubated in 37°C for 3 hours. Cells were then lysed in 50 ⁇ . of Glo reagent following manufacturer's instructions. Plates were read on Envision luminescent plate reader (Luminescent 96 well full area program). Percent ADCC activity was calculated as (Treated Cells- Spontaneous Cells)/(Total Lysis-Spontaneous Lysis)* 100.
  • DSC Differential Scanning Calorimetry
  • the IgGl-5m exhibits strikingly improved thermal stability when compared against CHO cell-culture derived IgG4PE (CHO IgG4PE) reference standard ( Figure 3B). As can be observed, all thermal transitions for the CHO IgG4PE material lie below 75°C, while the IgGl-5m is significantly stabilized and denatures at higher temperatures, with the exception of the CH2 domain at 62.2°C.
  • Example 10 The pharmacokinetics properties of anti-CD47 antibodies
  • the anti-CD47 antibodies IgG4-PE CHO, IgGl-CF and IgGl-5m-CF were administered by bolus intravenous injection to mice at dose levels of 3.0, 3.0 and 2.5 mg/kg, respectively.
  • Plasma samples were collectd at selected times out to 28 days (672 hours) after dosing, and the concentration of the respective protein determined by eimmunoassay.
  • the pharmacokinetic parameters were then calculated using a non-compartmental approach with WinNonlin V 5.3, Phoenix 64 (Certara, CA).
  • the AUC was calculated using the linear trapezoidal rule for the ascending portion of the curve and the log trapezoidal rule for the descending portion.
  • the terminal half-life was determined from a regression of the log of the plasma concentration versus time.
  • the number of points used for the regression was determined by visual inspection of the data using a minimum of three terminal time points.
  • the initial volume of distribution was calculated from the dose/plasma concentration extrapolated to zero time. All other parameters were calculated within WinNonlin using standard methods.
  • anti-tumor activity of anti-CD47 antibodies produced by the cell-free system were tested in vivo using a xenograft tumor model with the human myeloma cell line RPMI8226.
  • mice were injected subcutaneously with RPMI 8226 cells. Subsequently, mice were treated with vehicle control, hIgG4, or CF anti-CD47 antibodies, such as anti-CD47 IgGl-5m, were administered (qwx3) at a dose of 1 mg/kg, 0.3 mg/kg, or 0.1 mg/kg. Tumor volume were monitored.
  • Figure 5 depicts a graph of tumor volume versus days after tumor cell inoculation.
  • CF anti-CD47 IgGl -5m antibody achieved tumor volume reduction (TVR) of 83% at a dose of 1 mg/kg and a TVR of 50% at a dose of 0.3 mg/kg.
  • the percentage of tumor free mice at termination is 25% (2/8) for the 1 mg/kg dose of CF anti-CD47 IgGl -5m antibody.
EP15876093.4A 2014-12-30 2015-12-28 Anti-cd47-antikörper und verwendungen davon Pending EP3240569A4 (de)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462098291P 2014-12-30 2014-12-30
PCT/US2015/067642 WO2016109415A1 (en) 2014-12-30 2015-12-28 Anti-cd47 antibodies and uses thereof

Publications (2)

Publication Number Publication Date
EP3240569A1 true EP3240569A1 (de) 2017-11-08
EP3240569A4 EP3240569A4 (de) 2018-05-30

Family

ID=56284963

Family Applications (1)

Application Number Title Priority Date Filing Date
EP15876093.4A Pending EP3240569A4 (de) 2014-12-30 2015-12-28 Anti-cd47-antikörper und verwendungen davon

Country Status (17)

Country Link
US (2) US10870699B2 (de)
EP (1) EP3240569A4 (de)
JP (2) JP6850255B2 (de)
KR (1) KR102489471B1 (de)
CN (1) CN107530421B (de)
AU (1) AU2015374301B2 (de)
BR (1) BR112017014258A2 (de)
CA (1) CA2972604C (de)
CL (1) CL2017001736A1 (de)
CO (1) CO2017007673A2 (de)
EA (1) EA037654B1 (de)
EC (1) ECSP17041865A (de)
HK (1) HK1245154A1 (de)
MX (1) MX2017008819A (de)
SG (2) SG11201705310TA (de)
WO (1) WO2016109415A1 (de)
ZA (1) ZA201704467B (de)

Families Citing this family (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG11201705310TA (en) 2014-12-30 2017-07-28 Celgene Corp Anti-cd47 antibodies and uses thereof
AU2016258977C1 (en) * 2015-05-04 2022-07-14 Pieris Pharmaceuticals Gmbh Anti-cancer fusion polypeptide
US11427632B2 (en) * 2016-07-06 2022-08-30 Celgene Corporation Antibodies with low immunogenicity and uses thereof
CA3035681A1 (en) 2016-09-29 2018-04-05 Beijing Hanmi Pharmaceutical Co., Ltd. Heterodimeric immunoglobulin constructs and preparation methods thereof
JP7043074B2 (ja) 2016-10-20 2022-03-29 アイ-マブ バイオファーマ ユーエス リミテッド 新規なcd47モノクローナル抗体およびこの使用
EP3534965A4 (de) 2016-11-03 2020-06-24 Trillium Therapeutics Inc. Verbesserungen bei der cd47-blockadetherapie durch hdac-inhibitoren
EP3538557A4 (de) * 2016-11-08 2020-11-18 Ablexis, LLC Anti-cd47-antikörper
EP3533804A4 (de) 2016-11-18 2020-06-17 Beijing Hanmi Pharmaceutical Co., Ltd. Bispezifischer anti-pd-1/anti-her2- antikörper mit natürlicher antikörperstruktur und heterodimerer form und herstellungsverfahren dafür
WO2018137705A1 (en) * 2017-01-26 2018-08-02 Zai Lab (Shanghai) Co., Ltd. Cd47 antigen binding unit and uses thereof
WO2018152033A1 (en) * 2017-02-14 2018-08-23 Promab Biotechnologies, Inc. Cd47-car-t cells
CA3057841A1 (en) 2017-03-27 2018-10-04 Celgene Corporation Methods and compositions for reduction of immunogenicity
MX2019012295A (es) 2017-04-14 2020-02-07 Tollnine Inc Polinucleotidos inmunomoduladores, conjugados de anticuerpos de los mismos y metodos para su uso.
AU2018312222A1 (en) * 2017-08-02 2020-02-27 Phanes Therapeutics, Inc. Anti-CD47 antibodies and uses thereof
AU2018316742A1 (en) * 2017-08-18 2020-03-19 Centessa Pharmaceuticals (Uk) Limited Binding agents
US20210095019A1 (en) * 2017-11-10 2021-04-01 I-Mab Fusion Proteins Containing CD47 Antibodies and Cytokines
EP3717515A2 (de) 2017-12-01 2020-10-07 Seattle Genetics, Inc. Cd47 (maskierte)-antikörper und deren verwendung zur behandlung von krebs
JP7231641B2 (ja) * 2017-12-04 2023-03-01 北京韓美薬品有限公司 天然抗体様構造を有し、ヘテロ二量体形態の抗pd-l1/抗cd47二重特異性抗体、ならびにその製造方法
JP2021511064A (ja) * 2018-01-24 2021-05-06 ナンキン レジェンド バイオテック カンパニー リミテッド 重大な赤血球凝集を引き起こさない抗cd47抗体
WO2019153200A1 (zh) 2018-02-08 2019-08-15 北京韩美药品有限公司 抗pd-1/抗her2天然抗体结构样异源二聚体形式双特异抗体及其制备
GB201804860D0 (en) * 2018-03-27 2018-05-09 Ultrahuman Two Ltd CD47 Binding agents
CN110577597B (zh) * 2018-06-11 2021-10-22 康诺亚生物医药科技(成都)有限公司 一种阻断CD47和SIRPα相互作用的抗体
CN112566662A (zh) * 2018-06-15 2021-03-26 阿库鲁斯生物科学公司 针对cd47的阻断抗体及其使用方法
CN112601544A (zh) * 2018-07-05 2021-04-02 三钰生物科技股份有限公司 人类抗cd47抗体及其用途
CN110872348B (zh) * 2018-09-03 2021-09-03 长春金赛药业有限责任公司 人源化抗cd47单克隆抗体及其应用
AU2019370754A1 (en) * 2018-10-31 2021-02-04 I-Mab Biopharma Us Limited Novel CD47 antibodies and methods of using same
KR20210137477A (ko) * 2019-03-06 2021-11-17 지앙수 헨그루이 메디슨 컴퍼니 리미티드 이중기능성 융합 단백질 및 그것의 제약학적 용도
TW202104260A (zh) * 2019-04-05 2021-02-01 美商西建公司 腫瘤選擇性結合cd47之抗體之工程
US20200400662A1 (en) 2019-06-07 2020-12-24 ALX Oncology Inc. Methods and reagents for reducing the interference of drugs that bind cd47 in serological assays
KR20220047277A (ko) 2019-07-16 2022-04-15 길리애드 사이언시즈, 인코포레이티드 Hiv 백신, 및 이의 제조 및 사용 방법
AU2020365113A1 (en) 2019-10-18 2022-04-07 Forty Seven, Inc. Combination therapies for treating myelodysplastic syndromes and acute myeloid leukemia
CN114599681B (zh) * 2019-10-25 2024-01-09 上海药明生物技术有限公司 新型抗cd47抗体及其用途
MX2022005123A (es) 2019-10-31 2022-05-30 Forty Seven Inc Tratamiento basado en anti-cd47 y anti-cd20 para cancer hematologico.
CA3165735A1 (en) 2019-12-24 2021-07-01 Carna Biosciences, Inc. Diacylglycerol kinase modulating compounds
BR112022014623A2 (pt) 2020-02-14 2022-09-13 Jounce Therapeutics Inc Anticorpos e proteínas de fusão que se ligam a ccr8 e usos dos mesmos
MX2022010515A (es) 2020-02-28 2022-11-14 Tallac Therapeutics Inc Conjugacion mediada por transglutaminasa.
EP4168115A1 (de) 2020-06-22 2023-04-26 The Board of Trustees of the Leland Stanford Junior University Tsp-1-inhibitoren zur behandlung von gealtertem, atrophietem oder dystrophietem muskel
CA3192331A1 (en) * 2020-09-14 2022-03-17 Sutro Biopharma, Inc. Method for large scale production of antibodies using a cell-free protein synthesis system
CN116685348A (zh) 2020-10-07 2023-09-01 细胞基因公司 淋巴恶性肿瘤病状的双特异性抗体治疗
WO2022120286A1 (en) 2020-12-06 2022-06-09 ALX Oncology Inc. Multimers for reducing the interference of drugs that bind cd47 in serological assays
CN117209608A (zh) * 2020-12-23 2023-12-12 广东菲鹏制药股份有限公司 一种靶向cd47的抗体及其应用
EP4276113A1 (de) 2021-01-08 2023-11-15 Beijing Hanmi Pharmaceutical Co., Ltd. Spezifisch an cd47 bindender antikörper und antigenbindendes fragment davon
AU2022235341A1 (en) 2021-03-12 2023-09-21 Mendus B.V. Methods of vaccination and use of cd47 blockade
TW202302145A (zh) 2021-04-14 2023-01-16 美商基利科學股份有限公司 CD47/SIRPα結合及NEDD8活化酶E1調節次單元之共抑制以用於治療癌症
US20220389104A1 (en) * 2021-05-28 2022-12-08 Ose Immunotherapeutics Method for Treating CD127-Positive Cancers by Administering an Anti-CD127 Agent
CA3220923A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
AU2022298639A1 (en) 2021-06-23 2023-12-07 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
US11926628B2 (en) 2021-06-23 2024-03-12 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
US11976072B2 (en) 2021-06-23 2024-05-07 Gilead Sciences, Inc. Diacylglycerol kinase modulating compounds
WO2023076983A1 (en) 2021-10-28 2023-05-04 Gilead Sciences, Inc. Pyridizin-3(2h)-one derivatives
AU2022376954A1 (en) 2021-10-29 2024-05-02 Gilead Sciences, Inc. Cd73 compounds
WO2023122581A2 (en) 2021-12-22 2023-06-29 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
US20230242508A1 (en) 2021-12-22 2023-08-03 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
TW202340168A (zh) 2022-01-28 2023-10-16 美商基利科學股份有限公司 Parp7抑制劑
TW202346277A (zh) 2022-03-17 2023-12-01 美商基利科學股份有限公司 Ikaros鋅指家族降解劑及其用途
US20230355796A1 (en) 2022-03-24 2023-11-09 Gilead Sciences, Inc. Combination therapy for treating trop-2 expressing cancers
TW202345901A (zh) 2022-04-05 2023-12-01 美商基利科學股份有限公司 用於治療結腸直腸癌之組合療法
TW202400138A (zh) 2022-04-21 2024-01-01 美商基利科學股份有限公司 Kras g12d調節化合物
WO2024005423A1 (ko) * 2022-06-29 2024-01-04 고려대학교 산학협력단 인간 FCγRS 결합력이 제거된 당화 FC 변이체들
US20240116928A1 (en) 2022-07-01 2024-04-11 Gilead Sciences, Inc. Cd73 compounds
US20240010714A1 (en) * 2022-07-10 2024-01-11 Mbrace Therapeutics, Inc. Cell-free methods of recombinant antibody production
WO2024015741A1 (en) 2022-07-12 2024-01-18 Gilead Sciences, Inc. Hiv immunogenic polypeptides and vaccines and uses thereof
US20240091351A1 (en) 2022-09-21 2024-03-21 Gilead Sciences, Inc. FOCAL IONIZING RADIATION AND CD47/SIRPa DISRUPTION ANTICANCER COMBINATION THERAPY

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK1087013T3 (da) 1992-08-21 2009-05-11 Univ Bruxelles Immunoglobuliner uden lette kæder
US6005079A (en) 1992-08-21 1999-12-21 Vrije Universiteit Brussels Immunoglobulins devoid of light chains
DK0698097T3 (da) 1993-04-29 2001-10-08 Unilever Nv Produktion af antistoffer eller (funktionaliserede) fragmenter deraf afledt af Camelidae-immunoglobuliner med tung kæde
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
ES2331051T3 (es) 1999-11-29 2009-12-21 Bac Ip B.V. Inmovilizacion de moleculas de union de antigenos de un dominio.
WO2004006955A1 (en) 2001-07-12 2004-01-22 Jefferson Foote Super humanized antibodies
DE102007001370A1 (de) * 2007-01-09 2008-07-10 Curevac Gmbh RNA-kodierte Antikörper
MY149129A (en) * 2007-03-20 2013-07-15 Lilly Co Eli Anti-sclerostin antibodies
EP2111869A1 (de) 2008-04-23 2009-10-28 Stichting Sanquin Bloedvoorziening Zusammensetzungen und Verfahren zur Stärkung des Immunsystems
WO2011004847A1 (ja) 2009-07-07 2011-01-13 一般財団法人化学及血清療法研究所 新規ヒト化抗ヒトα9インテグリン抗体
PL2477648T3 (pl) 2009-09-15 2022-11-07 The Board Of Trustees Of The Leland Stanford Junior University Synergistyczna terapia anty-cd47 dla nowotworów hematologicznych
JP2014519338A (ja) * 2011-06-16 2014-08-14 ノバルティス アーゲー 治療薬として使用される可溶性タンパク質
HUE050875T2 (hu) * 2012-01-17 2021-01-28 Univ Leland Stanford Junior Nagy affinitású SIRP-alfa reagensek
US20140140989A1 (en) 2012-02-06 2014-05-22 Inhibrx Llc Non-Platelet Depleting and Non-Red Blood Cell Depleting CD47 Antibodies and Methods of Use Thereof
EA034778B1 (ru) 2012-02-06 2020-03-19 Инхибркс, Инк. Антитела к cd47 и способы их применения
SG11201506132PA (en) * 2013-02-06 2015-09-29 Inhibrx Llc Non-platelet depleting and non-red blood cell depleting cd47 antibodies and methods of use thereof
AU2014253785B2 (en) * 2013-04-19 2018-07-19 Sutro Biopharma, Inc. Expression of biologically active proteins in a bacterial cell-free synthesis system using cell extracts with elevated levels of exogenous chaperones
SG11201705310TA (en) 2014-12-30 2017-07-28 Celgene Corp Anti-cd47 antibodies and uses thereof
CN116425875A (zh) 2015-09-18 2023-07-14 安驰肿瘤公司 治疗性cd47抗体
JP2018535692A (ja) 2015-09-21 2018-12-06 エラスムス ユニバーシティ メディカル センターErasmus University Medical Center 抗cd47抗体及び使用方法

Also Published As

Publication number Publication date
JP6850255B2 (ja) 2021-03-31
HK1245154A1 (zh) 2018-08-24
EA037654B1 (ru) 2021-04-27
MX2017008819A (es) 2018-03-14
SG11201705310TA (en) 2017-07-28
KR20170100652A (ko) 2017-09-04
SG10202007176TA (en) 2020-08-28
JP2021048858A (ja) 2021-04-01
US20210054070A1 (en) 2021-02-25
CO2017007673A2 (es) 2018-01-05
CL2017001736A1 (es) 2018-03-16
EA201791485A1 (ru) 2018-02-28
JP2018506964A (ja) 2018-03-15
US20170369572A1 (en) 2017-12-28
KR102489471B1 (ko) 2023-01-18
ZA201704467B (en) 2019-09-25
AU2015374301B2 (en) 2021-02-11
CN107530421B (zh) 2021-07-20
US11787860B2 (en) 2023-10-17
AU2015374301A1 (en) 2017-07-20
WO2016109415A1 (en) 2016-07-07
US10870699B2 (en) 2020-12-22
EP3240569A4 (de) 2018-05-30
BR112017014258A2 (pt) 2018-03-06
ECSP17041865A (es) 2017-08-31
CN107530421A (zh) 2018-01-02
CA2972604C (en) 2023-07-04
CA2972604A1 (en) 2016-07-07

Similar Documents

Publication Publication Date Title
US11787860B2 (en) Anti-CD47 antibodies and uses thereof
AU2020202792B9 (en) Humanized or chimeric CD3 antibodies
WO2018089628A1 (en) Anti-ox40 antibodies, anti-gitr antibodies, and methods of use thereof
AU2017292752B2 (en) Antibodies with low immunogenicity and uses thereof
US20240075133A1 (en) Methods and compositions for reduction of immunogenicity
CA3177024A1 (en) Proteins binding nkg2d, cd16 and clec12a
CA3156983A1 (en) Antibodies against the poliovirus receptor (pvr) and uses thereof
CA3219672A1 (en) Anti-cea and anti-cd137 multispecific antibodies and methods of use
US11981745B2 (en) Anti-mesothelin antigen-binding molecules and uses thereof
US20230235091A1 (en) Anti-mesothelin antigen-binding molecules and uses thereof
NZ733261A (en) Anti-cd47 antibodies and uses thereof
WO2024044675A1 (en) Methods of cancer treatment using anti-pd1 antibodies in combination with anti-tim3 antibodies
EA045980B1 (ru) Антитела против рецептора полиовируса (pvr) и их применение
EA043164B1 (ru) Способы и композиции для снижения иммуногенности

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20170725

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
A4 Supplementary search report drawn up and despatched

Effective date: 20180502

RIC1 Information provided on ipc code assigned before grant

Ipc: C12P 21/08 20060101ALI20180424BHEP

Ipc: C07K 16/28 20060101ALI20180424BHEP

Ipc: A61K 39/395 20060101AFI20180424BHEP

Ipc: C12N 15/13 20060101ALI20180424BHEP

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1245154

Country of ref document: HK

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20190502

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

RAP3 Party data changed (applicant data changed or rights of an application transferred)

Owner name: CELGENE CORPORATION